1
|
Ioannou D, Tempest HG. The genetic basis of male and female infertility. Syst Biol Reprod Med 2025; 71:143-169. [PMID: 40294233 DOI: 10.1080/19396368.2025.2493621] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Revised: 04/04/2025] [Accepted: 04/10/2025] [Indexed: 04/30/2025]
Abstract
This review provides a comprehensive overview of the genetic factors underlying male and female infertility. Infertility affects an estimated one in six couples worldwide, with both male and female factors contributing equally to its prevalence. Approximately, 50% of infertility cases are attributed to genetic causes. We explore three main categories of genetic causes: chromosomal abnormalities, monogenic disorders, and syndromic conditions. Chromosomal causes, including numerical and structural aberrations, are discussed with a focus on their impact on gametogenesis and reproductive outcomes. We review key monogenic causes of infertility, highlighting recent discoveries in genes critical for gonadal development, gametogenesis, and hormonal regulation. Syndromic conditions affecting fertility are examined, highlighting their impact on reproductive function. Throughout the review, we address the challenges in identifying genetic mechanisms of infertility, particularly focusing on the intricate processes involved in oogenesis and spermatogenesis. We also discuss how advancements in genetic testing, such as next-generation sequencing (NGS) and genome-wide association studies (GWAS), have significantly enhanced our understanding of idiopathic infertility and promise further insights in the future. We also discuss the clinical implications of genetic diagnoses, including the role of preimplantation genetic testing (PGT) and genetic counseling in reproductive medicine. This review synthesizes current knowledge on the genetic basis of infertility, providing a comprehensive overview of chromosomal, monogenic, and syndromic causes. It aims to offer readers a solid foundation for understanding the complex genetic factors underlying reproductive disorders.
Collapse
Affiliation(s)
- Dimitrios Ioannou
- Department of Basic Sciences, College of Medicine, Roseman University of Health Sciences, Las Vegas, NV, USA
| | - Helen G Tempest
- Department of Basic Sciences, College of Medicine, Roseman University of Health Sciences, Las Vegas, NV, USA
| |
Collapse
|
2
|
Tummolo A, Paterno G, Carella R, Melpignano L, De Giovanni D. Exploring Partners, Parenting and Pregnancy Thinking in Late Adolescents and Young Adults with Inherited Metabolic Disorders. Pediatr Rep 2025; 17:56. [PMID: 40407581 PMCID: PMC12101304 DOI: 10.3390/pediatric17030056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/12/2025] [Revised: 04/13/2025] [Accepted: 04/29/2025] [Indexed: 05/26/2025] Open
Abstract
INTRODUCTION The psychosocial impact of living with an Inherited Metabolic Disorder (IMD) is becoming increasingly relevant and can have a significant impact on planning the future, conditioning the reproductive decisions made during adolescence and young adulthood. The aim of this paper is to explore thoughts about partner choices, parenthood and pregnancy among adolescents and young adults affected by IMDs. METHODS A cross-sectional study was performed. A sample of 23 patients affected by a range of IMDs were interviewed. Twenty-two questions were provided, distinguished into four main themes: partners, parenthood, pregnancy and need for information. RESULTS More than half of participants (57%) reported insecurities about relationships and declared that they were single for this reason, with most (70%) having a hope of having children in the future, although with the awareness and fear that they could also be affected. Almost all females (90%) consider themselves able to carry a pregnancy in a way similar to other women. There was the common need for more information about their potential fertility and parenthood linked to their condition. CONCLUSION Being diagnosed with an IMD can influence personal decisions regarding relationships and reproduction. The early identification of issues in these domains may enhance referrals for personalized interventions and build more focused support programmes.
Collapse
Affiliation(s)
- Albina Tummolo
- Department of Metabolic Diseases, Clinical Genetics and Diabetology, Giovanni XXIII Children Hospital, Azienda Ospedaliero-Universitaria Consorziale, 70126 Bari, Italy; (G.P.); (R.C.); (D.D.G.)
| | - Giulia Paterno
- Department of Metabolic Diseases, Clinical Genetics and Diabetology, Giovanni XXIII Children Hospital, Azienda Ospedaliero-Universitaria Consorziale, 70126 Bari, Italy; (G.P.); (R.C.); (D.D.G.)
| | - Rosa Carella
- Department of Metabolic Diseases, Clinical Genetics and Diabetology, Giovanni XXIII Children Hospital, Azienda Ospedaliero-Universitaria Consorziale, 70126 Bari, Italy; (G.P.); (R.C.); (D.D.G.)
| | - Livio Melpignano
- Medical Direction, Giovanni XXIII Children Hospital, Azienda Ospedaliero-Universitaria Consorziale, 70126 Bari, Italy;
| | - Donatella De Giovanni
- Department of Metabolic Diseases, Clinical Genetics and Diabetology, Giovanni XXIII Children Hospital, Azienda Ospedaliero-Universitaria Consorziale, 70126 Bari, Italy; (G.P.); (R.C.); (D.D.G.)
| |
Collapse
|
3
|
Hu C, Zhai L, Wang H. Knowledge, Attitudes, and Practices of the General Population Regarding Peripheral Blood Chromosomal Testing in the Premarital or Preconception Context. Mol Genet Genomic Med 2025; 13:e70103. [PMID: 40304124 PMCID: PMC12041940 DOI: 10.1002/mgg3.70103] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2024] [Revised: 04/06/2025] [Accepted: 04/15/2025] [Indexed: 05/02/2025] Open
Abstract
OBJECTIVES Some previous studies examined the knowledge, attitudes, and practices (KAP) toward prenatal genetic testing of the fetus but not toward blood chromosomal testing in the preconception and premarital period. This study investigated the KAP of the general population regarding peripheral blood chromosomal testing in the premarital or preconception period. DESIGN A cross-sectional study. SETTING From October 2023 to December 2023 at the authors' hospital. PARTICIPANTS Enrolled individuals who participated in free premarital medical examinations and free prepregnancy health examinations. PRIMARY AND SECONDARY OUTCOME MEASURES A self-designed questionnaire (Cronbach's α = 0.917) was used to collect the demographic information and KAP scores. KAP scores across variable categories were analyzed using the Mann-Whitney U test or Kruskal-Wallis H test. Correlations between KAP scores were evaluated by Pearson correlation analysis. Factors associated with KAP were identified by multivariable logistic regression. RESULTS This study included 630 valid questionnaires. The mean knowledge, attitude, and practice scores were 18.90 ± 4.48 (/30, 63.00%), 40.99 ± 5.32 (/50, 81.98%), and 37.66 ± 4.43 (/45, 83.69%), respectively. The knowledge scores were similar between genders (p = 0.840). Compared with males, females had higher attitude scores (42.01 ± 5.40 vs. 40.01 ± 5.07, p < 0.001) and higher practice scores (38.17 ± 4.44 vs. 37.18 ± 4.37, p = 0.013). There were no significant differences between genders regarding the frequencies of having undergone chromosomal testing and recommending it to the partner. The knowledge scores (OR = 1.060, 95% CI: 1.018-1.103, p = 0.004) and attitude scores (OR = 1.198, 95% CI: 1.154-1.244, p < 0.001) were positively independently associated with the practice scores. CONCLUSIONS The general population in Hangzhou displays poor knowledge but favorable attitudes and proactive practices regarding peripheral blood chromosomal testing in the premarital or preconception period. Cultivating proper knowledge and attitude should improve practice.
Collapse
Affiliation(s)
- Caixia Hu
- Hangzhou Linping District Maternal & Child Health Care HospitalHangzhouChina
| | - Lulu Zhai
- Hangzhou Linping District Maternal & Child Health Care HospitalHangzhouChina
| | - Hailian Wang
- Hangzhou Linping District Maternal & Child Health Care HospitalHangzhouChina
| |
Collapse
|
4
|
Alibakhshi R, Soleimani M, Yari K, Kalhori AA, Karami M, Kalhori MR. Role of miRNA polymorphisms on male and female infertility and recurrent implantation failure. J Obstet Gynaecol Res 2025; 51:e16281. [PMID: 40174631 DOI: 10.1111/jog.16281] [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: 09/11/2024] [Accepted: 03/20/2025] [Indexed: 04/04/2025]
Abstract
PURPOSE MiRNAs regulate gene expression, impacting reproductive health, such as infertility and implantation failure. This study investigated the association between miRNA polymorphisms and male infertility (azoospermia, oligospermia), female infertility (endometriosis), and recurrent implantation failure in IVF. METHODS In order to find relevant articles, a search was conducted in PubMed, WOS, Scopus, and the Cochrane Library databases using Mesh terms and Entry terms with the keywords (miRNA OR microRNA polymorphism) AND (abortion OR miscarriage OR infertility OR sterility OR pregnancy loss OR implantation failure). RESULTS The study revealed miRNA polymorphisms linked to a higher risk of male and female infertility and repeated implantation failure in IVF. These results underscore the significance of genetic variations in miRNAs in developing reproductive issues and the promise of personalized treatment strategies guided by genetic profiles. CONCLUSION These results provide valuable insights into the genetic basis of infertility and emphasize the need for further research to develop targeted diagnostic tools and therapies. Understanding the role of miRNA polymorphisms in reproductive health has significant implications for improving outcomes for couples struggling with infertility. By elucidating the genetic factors contributing to infertility, this study paves the way for personalized approaches to diagnosis and treatment, ultimately enhancing reproductive care.
Collapse
Affiliation(s)
- Reza Alibakhshi
- Department of Biochemistry, School of Medicine, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Masoud Soleimani
- Department of Hematology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Kheirollah Yari
- Medical Biology Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Amir Ali Kalhori
- Student Research Committee, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Mohsen Karami
- Clinical Research Development Center, Imam Khomeini and Mohammad Kermanshahi and Farabi Hospitals, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Mohammad Reza Kalhori
- Regenerative Medicine Research Center, Kermanshah University of Medical Sciences, Kermanshah, Iran
- Fertility and Infertility Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| |
Collapse
|
5
|
Yang N, Wei XG, Li K, Wang F, Song F, Sun W, Wang Y, Zhao Z, Mu J, Ma H. Transcriptomic analysis of the HPT axis in a model of oligoasthenozoospermia induced by Adenine in rats. Exp Mol Pathol 2025; 141:104948. [PMID: 39700678 DOI: 10.1016/j.yexmp.2024.104948] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2023] [Revised: 11/25/2024] [Accepted: 12/11/2024] [Indexed: 12/21/2024]
Abstract
Male infertility is most commonly caused by oligozoospermia, and its pathogenesis is still poorly understood at the molecular level. This study used RNA sequencing (RNA-Seq) technology to identify candidate genes and regulatory pathways that regulate semen quality in the hypothalamic, pituitary, and testicular tissues of healthy rats and Adenine-induced oligozoospermia model rats. Semen quality testing and histological analysis of testicular tissues were performed on both groups of rats. We identified 627, 692, and 437 differentially expressed genes in the hypothalamus, pituitary gland, and testes, respectively. Functional analysis indicates that "neuronal projections," "positive regulation of hormone biosynthetic process," and "neuroactive ligand-receptor interaction pathways" are closely related to the hypothalamic-pituitary-testicular (HPT) axis hormone regulation and sperm production. Seven genes (Pomc, Rxfp1, Tac1, Npy, Insl3, Hsd3b3, Lhcgr) have been identified as key candidate genes responsible for regulating sperm quality within the HPT axis, potentially affecting rat reproductive function by influencing testicular development and testosterone secretion. These data provide a theoretical basis for further understanding the molecular mechanisms of reproductive performance in a rat model of oligoasthenozoospermia.
Collapse
Affiliation(s)
- Nan Yang
- College of Traditional Chinese Medicine, Ningxia Medical University, Yinchuan 750000, China; Key Laboratory of Modernization of Minority Medicine, Ministry of Education, Ningxia Medical University, Yinchuan 750000, China
| | - Xiao-Ge Wei
- College of Traditional Chinese Medicine, Ningxia Medical University, Yinchuan 750000, China; Key Laboratory of Modernization of Minority Medicine, Ministry of Education, Ningxia Medical University, Yinchuan 750000, China
| | - Kaiying Li
- College of Traditional Chinese Medicine, Ningxia Medical University, Yinchuan 750000, China; Key Laboratory of Modernization of Minority Medicine, Ministry of Education, Ningxia Medical University, Yinchuan 750000, China
| | - Fei Wang
- College of Traditional Chinese Medicine, Ningxia Medical University, Yinchuan 750000, China; Key Laboratory of Modernization of Minority Medicine, Ministry of Education, Ningxia Medical University, Yinchuan 750000, China
| | - Fei Song
- Lianyungang No. 1 People's Hospital, Lianyungang 222000, Jiangsu Province, China
| | - Wenjing Sun
- College of Traditional Chinese Medicine, Ningxia Medical University, Yinchuan 750000, China; Key Laboratory of Modernization of Minority Medicine, Ministry of Education, Ningxia Medical University, Yinchuan 750000, China
| | - Yan Wang
- College of Traditional Chinese Medicine, Ningxia Medical University, Yinchuan 750000, China
| | - Zhenning Zhao
- College of Traditional Chinese Medicine, Ningxia Medical University, Yinchuan 750000, China
| | - Jing Mu
- College of Traditional Chinese Medicine, Ningxia Medical University, Yinchuan 750000, China.
| | - Huisheng Ma
- College of Traditional Chinese Medicine, Ningxia Medical University, Yinchuan 750000, China; Key Laboratory of Modernization of Minority Medicine, Ministry of Education, Ningxia Medical University, Yinchuan 750000, China.
| |
Collapse
|
6
|
Farahani M, Jalali A, Moghadasi S, Rezaei M, Khodadadi R. Investigating the Impact of Bismuth Oxide Nanoparticles on Dazl Gene Expression and Spermatogenesis Indices in Male Mice. Biol Trace Elem Res 2025:10.1007/s12011-025-04554-9. [PMID: 40011410 DOI: 10.1007/s12011-025-04554-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/14/2025] [Accepted: 02/15/2025] [Indexed: 02/28/2025]
Abstract
Bismuth (Bi) exposure has been linked to various health effects, but its direct impact on male fertility remains largely unexplored. This study investigated the effects of bismuth oxide nanoparticles (Bi₂O₃ NPs) on male reproductive function in Naval Medical Research Institute (NMRI) mice. The mice were randomly allocated to seven groups: one control receiving physiological saline and six treatment groups receiving Bi₂O₃ NPs (25, 50, 100, 200, 400, and 800 mg/kg body weight/day) for 35 days. Treatment was administered via oral gavage. Following treatment, we evaluated body weight, blood serum biochemistry, Dazl gene expression, sperm quality, testicular histology, and cell counts (spermatogenic, Leydig, and Sertoli cells). Compared with the control, Bi₂O₃ NPs exposure resulted in significant reductions in testosterone levels, total antioxidant capacity, superoxide dismutase activity, Dazl gene expression, sperm motility, count, viability, normal morphology, and DNA integrity, and the level of malondialdehyde (MDA) increased. Additionally, testicular tissue and seminiferous tubule volume decreased, whereas interstitial tissue volume increased. Notably, sperm production was impaired, as evidenced by reduced numbers of spermatogonia, spermatocytes, spermatids, Sertoli cells, and Leydig cells. Body weight and seminiferous tubule basement membrane parameters remained largely unaffected. These findings suggest that Bi₂O₃ NPs induce oxidative stress, leading to lipid peroxidation and ultimately compromising male fertility. Our study highlights the potential detrimental effects of Bi₂O₃ NPs exposure on male reproductive health and warrants further investigation into their impact on human fertility at relevant concentrations.
Collapse
Affiliation(s)
- Mohadese Farahani
- Department of Biology, Faculty of Science, Arak University, Arak, 384817758, Iran
| | - Amir Jalali
- Department of Biology, Faculty of Science, Arak University, Arak, 384817758, Iran.
| | - Samira Moghadasi
- Department of Biology, Faculty of Science, Shahed University, Tehran, Iran
| | - Marziyeh Rezaei
- Department of Cell and Molecular Biology and Microbiology, Faculty of Science, University of Isfahan, Isfahan, Iran
| | - Reyhaneh Khodadadi
- Department of Biology, Faculty of Science, Arak University, Arak, 384817758, Iran
| |
Collapse
|
7
|
Jašinskienė E, Sniečkutė I, Galminas I, Žemaitis L, Simutis M, Čaplinskienė M. Evaluation of Risk Factors and a Gene Panel as a Tool for Unexplained Infertility Diagnosis by Next-Generation Sequencing. MEDICINA (KAUNAS, LITHUANIA) 2025; 61:271. [PMID: 40005388 PMCID: PMC11857236 DOI: 10.3390/medicina61020271] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 12/15/2024] [Revised: 01/22/2025] [Accepted: 01/31/2025] [Indexed: 02/27/2025]
Abstract
Background and Objective: Unexplained infertility is a major challenge in reproductive medicine and requires advanced diagnostic approaches to identify the underlying factors accurately. This study aims to evaluate the utility of risk factor analysis and a gene panel in diagnosing unexplained infertility using the next-generation sequencing (NGS) technology. Our study aimed to characterize and identify risk and genetic factors associated with unexplained infertility. Materials and methods: A cohort of patients with unexplained infertility was comprehensively screened for risk factors and genetic variations using a targeted gene panel (10 couples with unexplained infertility (UI) and 36 fertile couples). 108 articles were selected (58 on female infertility and 50 on male infertility) presenting genes that may be associated with unexplained infertility. A gene panel for unexplained infertility was compiled based on the literature data. A customized virtual panel was created from the exome sequencing data. Results: In the female group, controls had a higher mean age, while in the male patients, both groups were similar in terms of age. Both gender groups had comparable BMI values. No significant associations (p > 0.05) between risk factors and unexplained infertility were found when evaluating anthropometric parameters and other sociodemographic characteristics. In two male patients (20%), a molecular defect was detected in NGS variants classified aspossible benign and probably benign In particular, missense variants were identified in the UGT2B7 and CATSPER2 genes, A molecular defect classified as probably damaging was found in five female patients (50%). In particular, missense variants were identified in the CAPN10, MLH3, HABP2, IRS1, GDF9, and SLC19A1 genes. Conclusions: The study emphasizes that unexplained infertility is often related to mechanisms beyond causative mutations and highlights the need for integrative genomic research involving broader gene panels and multi-faceted approaches, including transcriptomics and epigenetics, to uncover latent genetic predispositions.
Collapse
Affiliation(s)
- Eglė Jašinskienė
- Department of Biochemistry, Vytautas Magnus University, K. Donelaicio St. 58, 44248 Kaunas, Lithuania;
| | - Ieva Sniečkutė
- UAB Genomika, K. Barsausko St. 59, 51423 Kaunas, Lithuania; (I.S.); (I.G.); (L.Ž.); (M.S.)
| | - Ignas Galminas
- UAB Genomika, K. Barsausko St. 59, 51423 Kaunas, Lithuania; (I.S.); (I.G.); (L.Ž.); (M.S.)
| | - Lukas Žemaitis
- UAB Genomika, K. Barsausko St. 59, 51423 Kaunas, Lithuania; (I.S.); (I.G.); (L.Ž.); (M.S.)
| | - Mantas Simutis
- UAB Genomika, K. Barsausko St. 59, 51423 Kaunas, Lithuania; (I.S.); (I.G.); (L.Ž.); (M.S.)
| | - Marija Čaplinskienė
- Department of Biochemistry, Vytautas Magnus University, K. Donelaicio St. 58, 44248 Kaunas, Lithuania;
| |
Collapse
|
8
|
Chian R, Guan Y, He X, Xu J, Shu J, Li J. The quality of human eggs and its pre-IVF incubation. Reprod Med Biol 2025; 24:e12652. [PMID: 40321658 PMCID: PMC12048747 DOI: 10.1002/rmb2.12652] [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: 03/03/2025] [Accepted: 04/08/2025] [Indexed: 05/08/2025] Open
Abstract
Background Multi-factors influence the success rate of infertility treatments, and one of the important points is to obtain good quality eggs. Methods Based on the literatures and unpublished data, the factors affecting egg quality were summarized. Main Findings Results Egg quality is an important determinant in successful infertility treatment. In addition to maternal age, controlled ovarian hyperstimulation (COH) protocols also play a key role in affecting the quality of the egg. After egg retrieval, the insemination occurs 3-6 h after collection, with a pre-IVF incubation time by in vitro fertilization (IVF) or intracytoplasmic sperm injection (ICSI) (39-42 h post-HCG injection). The pre-IVF incubation refers to the short period time of 3 to 6 h after oocyte retrieval and before the insemination by IVF or ICSI. The pre-IVF incubation of collected eggs in the designed culture medium improves egg quality in terms of maturation and early embryonic development. Conclusions Pre-IVF incubation of the collected eggs contributes to the improvement of the quality of eggs; therefore, it may increase subsequent pregnancy and implantation rates following embryo transfer.
Collapse
Affiliation(s)
- Ri‐Cheng Chian
- Laboratory of Research and DevelopmentARSCI Biomedical Inc.Jiaxing CityPeople's Republic of China
| | - Yi‐Chun Guan
- Center for Reproductive MedicineThe Third Affiliated Hospital of Zhengzhou UniversityZhengzhou CityPeople's Republic of China
| | - Xiao‐Jin He
- Center for Reproductive MedicineThe First People's Hospital of Jiaotong UniversityShanghaiPeople's Republic of China
| | - Jian Xu
- Center for Reproductive MedicineThe Fourth Affiliated Hospital, Zhejiang University School of MedicineYiwuPeople's Republic of China
| | - Jin‐Hui Shu
- Center of Reproductive MedicineMaternal and Child Health Hospital of Guangxi Zhuang Autonomous RegionNanningPeople's Republic of China
| | - Jian‐Hua Li
- Reproductive Medical Center, Department of Obstetrics and GynecologySeventh Medical Center of PLA General HospitalBeijingPeople's Republic of China
| |
Collapse
|
9
|
Ikbal Atli E, Yalcintepe S, Atli E, Demir S, Gurkan H. Next-Generation Sequencing Infertility Panel in Turkey: First Results. Balkan J Med Genet 2024; 27:49-57. [PMID: 40070865 PMCID: PMC11892938 DOI: 10.2478/bjmg-2024-0019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/14/2025] Open
Abstract
Background Male infertility is a complex pathophysiological disorder. At least 2000 genes are implicated in the etiology of male infertility, making it a very complex genetic condition. In cases of male infertility, genetic testing using next-generation sequencing (NGS) technology may be useful for diagnosis. Thus, the purpose of this investigation was to apply the diagnostic offer for genetic variant identification using an NGS panel. Methods We developed an NGS gene panel that we used in 85 infertile male patients. The panel consisted of 132 genes exploring the genetic causes of male infertility; namely spermatogenesis failure due to single-gene mutations, central hypogonadism, androgen insensitivity syndrome, congenital hypopituitarism, and primary ciliary dyskinesia etc. Results A total of 85 patients (85 males) between 21 year and 45 years old were included in the study group. NGS analysis had been applied in all the primary infertility cases. As a result of NGS analysis, 58 clinical variants in 28 genes were detected in 41 patients (%48.23- 41/85). Conclusion Consequently, pre-diagnostic genes included in a custom-made NGS panel test can enhance genetic diagnostic testing and have an impact on the clinical management of male infertility.
Collapse
Affiliation(s)
- E Ikbal Atli
- Trakya University, Faculty of Medicine, Department of Medical Genetics, Edirne, Turkey
| | - S Yalcintepe
- Trakya University, Faculty of Medicine, Department of Medical Genetics, Edirne, Turkey
| | - E Atli
- Trakya University, Faculty of Medicine, Department of Medical Genetics, Edirne, Turkey
| | - S Demir
- Trakya University, Faculty of Medicine, Department of Medical Genetics, Edirne, Turkey
| | - H Gurkan
- Trakya University, Faculty of Medicine, Department of Medical Genetics, Edirne, Turkey
| |
Collapse
|
10
|
Benonisdottir S, Straub VJ, Kong A, Mills MC. Genetics of female and male reproductive traits and their relationship with health, longevity and consequences for offspring. NATURE AGING 2024; 4:1745-1759. [PMID: 39672892 DOI: 10.1038/s43587-024-00733-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Accepted: 09/26/2024] [Indexed: 12/15/2024]
Abstract
Substantial shifts in reproductive behaviors have recently taken place in many high-income countries including earlier age at menarche, advanced age at childbearing, rising childlessness and a lower number of children. As reproduction shifts to later ages, genetic factors may become increasingly important. Although monogenic genetic effects are known, the genetics underlying human reproductive traits are complex, with both causal effects and statistical bias often confounded by socioeconomic factors. Here, we review genome-wide association studies (GWASs) of 44 reproductive traits of both female and male individuals from 2007 to early 2024, examining reproductive behavior, reproductive lifespan and aging, infertility and hormonal concentration. Using the GWAS Catalog as a basis, from 159 relevant studies, we isolate 37 genes that harbor association signals for four or more reproductive traits, more than half of which are linked to rare Mendelian disorders, including ten genes linked to reproductive-related disorders: FSHB, MCM8, DNAH2, WNT4, ESR1, IGSF1, THRB, BRWD1, CYP19A1 and PTPRF. We also review the relationship of reproductive genetics to related health and behavioral traits, aging and longevity and the effect of parental age on offspring outcomes as well as reflecting on limitations, open questions and challenges in this fast-moving field.
Collapse
Affiliation(s)
- Stefania Benonisdottir
- Leverhulme Centre for Demographic Science, Nuffield Department of Population Health, University of Oxford and Nuffield College, Oxford, UK
- Institute of Physical Science, University of Iceland, Reykjavik, Iceland
- Faculty of Medicine, University of Iceland, Reykjavik, Iceland
| | - Vincent J Straub
- Leverhulme Centre for Demographic Science, Nuffield Department of Population Health, University of Oxford and Nuffield College, Oxford, UK
| | - Augustine Kong
- Leverhulme Centre for Demographic Science, Nuffield Department of Population Health, University of Oxford and Nuffield College, Oxford, UK
| | - Melinda C Mills
- Leverhulme Centre for Demographic Science, Nuffield Department of Population Health, University of Oxford and Nuffield College, Oxford, UK.
- Department of Genetics, University Medical Centre Groningen, Groningen, the Netherlands.
- Department of Economics, Econometrics and Finance, University of Groningen, Groningen, the Netherlands.
| |
Collapse
|
11
|
Khashei Varnamkhasti K, Khashei Varnamkhasti S, Bahraini N, Davoodi M, Sadeghian M, Khavanin M, Naeimi R, Naeimi S. Multi-locus high-risk alleles association from interleukin's genes with female infertility and certain comorbidities. BMC Res Notes 2024; 17:344. [PMID: 39580416 PMCID: PMC11585211 DOI: 10.1186/s13104-024-06988-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Accepted: 10/28/2024] [Indexed: 11/25/2024] Open
Abstract
Objective There is evidence that cytokine genes' single nucleotide polymorphisms could be the reasons behind female infertility. This study aimed to identify the role for Interleukin33 rs1048274 (G > A) and rs16924243 (T > C), Interleukin22 rs1397852121 (C > T), rs1295978671 (C > T) and rs2227483 (A > T), Interleukin17A rs2275913 (G > A,C) and Interleukin17F rs763780 (T > C), Interleukin13 1512 (A > C) and IL13 2044 (G > A), and Interleukin4 rs2243250 (C > T) and rs2070874 (C > T) gene polymorphisms in female infertility to gain a richly more detailed understanding of its genetic predisposition. Five distinct groups, each comprising 200 infertile women and 200 age-matched fertile controls, were recruited to each Interleukins (33, 22, 17, 13 and 4) in this case-control study and were genotyped by using an amplification refractory mutation system. Statistical analysis is conducted by SPSS software V. 22 and using Chi-square (χ2) and logistic regression tests. Strength of association was estimated by multiple-comparison correction, population structure test and Haplotype analysis. The study was approved by the Academic Ethics Committee and each enrolled patient signed an informed consent.Results Our statistical results revealed risk alleles in all of the substitution lines for women infertility. Current findings provided evidence that in the presence of Interleukin33 Ap-value rs1048274 = 0.002 and Cp-value rs16924243 < 0.0001, Interleukin 22Tp-value rs1397852121 < 0.0001 and Tp-value rs2227483 = 0.000, Interleukin17A Ap-value rs2275913 = 0.003 and Interleukin17F Cp-value rs763780 = 0.000 and Interleukin13 Cp-value 1512 = 0.000 and Ap-value 2044 = 0.003, Interleukin4 Tp-value rs2243250 = 0.001 and Tp-value rs2070874 = 0.009 risk alleles, risk genotype also were significantly associated with increased chances of developing infertility. The relationship between risk genotypes and several well-established infertility risk factors including, polycystic ovary syndrome, premature ovarian failure, oophorectomy, diminished ovarian reserve, endometriosis, uterine fibroids, ovarian cysts, uterine polyps, fallopian tube blockage and thyroid dysfunction, also exhibited. This study suggests the significant role of interleukin gene polymorphisms in human reproductive success.
Collapse
Affiliation(s)
- Khalil Khashei Varnamkhasti
- Department of Medical Laboratory Sciences, Faculty of Medicine, Kazerun Branch, Islamic Azad University, Kazerun, Iran
| | - Samire Khashei Varnamkhasti
- Department of Medical Laboratory Sciences, Faculty of Medicine, Kazerun Branch, Islamic Azad University, Kazerun, Iran
| | - Najmeh Bahraini
- Department of Genetics, College of Science, Kazerun Branch, Islamic Azad University, Kazerun, Iran
| | - Mohaddeseh Davoodi
- Department of Genetics, College of Science, Kazerun Branch, Islamic Azad University, Kazerun, Iran
| | - Mahsa Sadeghian
- Department of Genetics, College of Science, Kazerun Branch, Islamic Azad University, Kazerun, Iran
| | - Massomeh Khavanin
- Department of Genetics, College of Science, Kazerun Branch, Islamic Azad University, Kazerun, Iran
| | - Raana Naeimi
- Department of Genetics, College of Science, Kazerun Branch, Islamic Azad University, Kazerun, Iran
| | - Sirous Naeimi
- Department of Biology, Zand Institute of Higher Education, Shiraz, Iran.
| |
Collapse
|
12
|
Martinez V, Yen IH, Alvarez C, Williams AD, Ha S. Exposure to Environmental Chemicals and Infertility Among US Reproductive-Aged Women. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2024; 21:1541. [PMID: 39767383 PMCID: PMC11675402 DOI: 10.3390/ijerph21121541] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Revised: 11/14/2024] [Accepted: 11/19/2024] [Indexed: 01/11/2025]
Abstract
Environmental chemical exposure has been rising over the past few decades but its impact on fertility remains uncertain. We assessed exposures to 23 common chemicals across a range of sociodemographic characteristics and their relationship with self-reported infertility. The analytic sample was non-pregnant women aged 18-49 years without a history of hysterectomy or oophorectomy (n = 2579) from the National Health and Nutrition Examination Survey (2013-2016). Environmental chemical exposure was assessed with biospecimens and dichotomized as high and low levels of exposure based on the median. Logistic regression models estimated the adjusted odds ratio (aOR) and 95% confidence intervals (CIs) for the association between high levels of exposure and infertility, adjusted for age, race, education level, family income, and smoking status. We observed associations between infertility and cadmium [aOR: 1.88; 95% CI: 1.02-3.47] and arsenic [aOR: 1.88 (1.05-3.36)]. Two pesticides hexachlorobenzene [OR: 2.04 (1.05-3.98)] and oxychlordane [OR: 2.04 (1.12-3.69)] were also associated with infertility in unadjusted analyses. There were negative associations with two Per- and polyfluoroalkyl substances with n-perfluorooctanoic acid [aOR: 0.51: (0.30-0.86)] and n-perfluorooctane sulfonic acid [aOR: 0.51: (0.26-0.97). Specific chemicals may contribute to infertility risk, highlighting the need for targeted public health strategies to mitigate exposure.
Collapse
Affiliation(s)
- Valerie Martinez
- Public Health Department, School of Social Sciences, Humanities and Arts, Health Science Research Institute, University of California, Merced, CA 95343, USA; (V.M.); (I.H.Y.)
| | - Irene H. Yen
- Public Health Department, School of Social Sciences, Humanities and Arts, Health Science Research Institute, University of California, Merced, CA 95343, USA; (V.M.); (I.H.Y.)
| | - Camila Alvarez
- Department of Sociology, University of California, San Diego, CA 92122, USA;
| | - Andrew D. Williams
- Public Health Program, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, ND 58202, USA;
| | - Sandie Ha
- Public Health Department, School of Social Sciences, Humanities and Arts, Health Science Research Institute, University of California, Merced, CA 95343, USA; (V.M.); (I.H.Y.)
| |
Collapse
|
13
|
Arora M, Mehta P, Sethi S, Anifandis G, Samara M, Singh R. Genetic etiological spectrum of sperm morphological abnormalities. J Assist Reprod Genet 2024; 41:2877-2929. [PMID: 39417902 PMCID: PMC11621285 DOI: 10.1007/s10815-024-03274-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2024] [Accepted: 09/18/2024] [Indexed: 10/19/2024] Open
Abstract
PURPOSE Male infertility manifests in the form of a reduction in sperm count, sperm motility, or the loss of fertilizing ability. While the loss of sperm production can have mixed reasons, sperm structural defects, cumulatively known as teratozoospermia, have predominantly genetic bases. The aim of the present review is to undertake a comprehensive analysis of the genetic mutations leading to sperm morphological deformities/teratozoospermia. METHODS We undertook literature review for genes involved in sperm morphological abnormalities. The genes were classified according to the type of sperm defects they cause and on the basis of the level of evidence determined by the number of human studies and the availability of a mouse knockout. RESULTS Mutations in the SUN5, CEP112, BRDT, DNAH6, PMFBP1, TSGA10, and SPATA20 genes result in acephalic sperm; mutations in the DPY19L2, SPATA16, PICK1, CCNB3, CHPT1, PIWIL4, and TDRD9 genes cause globozoospermia; mutations in the AURKC gene cause macrozoospermia; mutations in the WDR12 gene cause tapered sperm head; mutations in the RNF220 and ADCY10 genes result in small sperm head; mutations in the AMZ2 gene lead to vacuolated head formation; mutations in the CC2D1B and KIAA1210 genes lead to pyriform head formation; mutations in the SEPT14, ZPBP1, FBXO43, ZCWPW1, KATNAL2, PNLDC1, and CCIN genes cause amorphous head; mutations in the SEPT12, RBMX, and ACTL7A genes cause deformed acrosome formation; mutations in the DNAH1, DNAH2, DNAH6, DNAH17, FSIP2, CFAP43, AK7, CHAP251, CFAP65, ARMC2 and several other genes result in multiple morphological abnormalities of sperm flagella (MMAF). CONCLUSIONS Altogether, mutations in 31 genes have been reported to cause head defects and mutations in 62 genes are known to cause sperm tail defects.
Collapse
Affiliation(s)
- Manvi Arora
- Division of Endocrinology, CSIR-Central Drug Research Institute, Lucknow, India
| | - Poonam Mehta
- Division of Endocrinology, CSIR-Central Drug Research Institute, Lucknow, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Shruti Sethi
- Division of Endocrinology, CSIR-Central Drug Research Institute, Lucknow, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - George Anifandis
- Department of Obstetrics and Gynaecology, School of Health Sciences, Faculty of Medicine, University of Thessaly, Larisa, Greece
| | - Mary Samara
- Department of Obstetrics and Gynaecology, School of Health Sciences, Faculty of Medicine, University of Thessaly, Larisa, Greece
| | - Rajender Singh
- Division of Endocrinology, CSIR-Central Drug Research Institute, Lucknow, India.
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India.
| |
Collapse
|
14
|
Liu K, Liu Y, Chu M. Detection of polymorphisms in six genes and their association analysis with litter size in sheep. Anim Biotechnol 2024; 35:2309954. [PMID: 38294691 DOI: 10.1080/10495398.2024.2309954] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2024]
Abstract
Litter size in sheep is a complex trait controlled by micro-effective polygenes. APAF1, CLSTN2, CTH, PLCB1, PLCB4, and CHST11 are all involved in mammalian reproduction. However, the effects of these genes on litter size in sheep are still unclear. Therefore, in this study, we used Sequenom MassARRAY® SNP assay technology to type the single nucleotide polymorphisms (SNPs) loci of six genes in five sheep breeds. The results showed that most sheep breeds contain three genotypes at each locus. Then, we conducted population genetic analysis on the SNPs of six genes and found that the polymorphic information content in all sheep breeds ranged from 0 to 0.37, and most sheep breeds were in Hardy-Weinberg equilibrium (p > 0.05). In addition, association analysis in Small Tail Han sheep indicated that the rs399534524 locus in CLSTN2 was highly associated with first parity litter size, and litter size in ewes with CT genotype was higher than that in ewes with CC genotype or TT genotype. Furthermore, the rs407142552 locus in CTH was highly associated with second parity litter size in Small Tail Han sheep, and litter size in ewes with CT genotype was higher than that in ewes with TT genotype. Finally, we predicted the CTH and CLSTN2 protein interaction network and found that HTR1E, NOM1, CCDC174 and ALPK3 interact with CLSTN2 and have been reported as candidate genes related to litter size in sheep. These results suggest that they may be useful genetic markers for increasing litter size in sheep.
Collapse
Affiliation(s)
- Kai Liu
- State Key Laboratory of Animal Biotech Breeding, Institute of Animal Science, Chinese Academy of Agricultural Sciences (CAAS), Beijing, China
| | - Yufang Liu
- State Key Laboratory of Animal Biotech Breeding, Institute of Animal Science, Chinese Academy of Agricultural Sciences (CAAS), Beijing, China
| | - Mingxing Chu
- State Key Laboratory of Animal Biotech Breeding, Institute of Animal Science, Chinese Academy of Agricultural Sciences (CAAS), Beijing, China
| |
Collapse
|
15
|
Farag MR, El-Naseery NI, El Behery EI, Nouh DS, El-Mleeh A, Mostafa IMA, Alagawany M, Di Cerbo A, Azzam MM, Mawed SA. The Role of Chlorella vulgaris in Attenuating Infertility Induced by Cadmium Chloride via Suppressing Oxidative Stress and Modulating Spermatogenesis and Steroidogenesis in Male Rats. Biol Trace Elem Res 2024; 202:4007-4020. [PMID: 38114777 DOI: 10.1007/s12011-023-03971-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Accepted: 11/20/2023] [Indexed: 12/21/2023]
Abstract
Cadmium (Cd) is an environmental pollutant known as endocrine disruptor . Cd has been reported to induce perturbations of the testicular functions and the subsequent decline of the male fertility of both animals and humans. Chlorella vulgaris (ChV) a species of green microalga has been reported to have multiple beneficial activities such as anti-inflammatory, antioxidant, and antiapoptotic effects. Thus, this work was conducted to declare the benefits of Chlorella vulgaris (ChV) (500 mg/kg doses) against cadmium chloride CdCl2 (2 mg/kg doses) toxicity on the main and accessory reproductive organs' weight, structure, and function of male rats. Briefly, 40 adult male rats in 4 groups (n = 10) were used as follows; control, ChV, CdCl2, and CdCl2+ChV. (i) The 1st group was kept as control fed on pellet chow and water ad libitum. (ii) The second group is Chlorella vulgaris (ChV) group fed with C. vulgaris alga for 10 days (500 mg/kg BW). (iii) The third group was administrated CdCl2 (2mg/kg BW) via subcutaneous injection (S/C) daily for 10 days. (iv) The fourth group administered both CdCl2 and ChV with the abovementioned doses daily for successive 10 days. Our observations declared that cadmium exhibited an adverse influence on the testes and prostate gland architecture indicated by seminiferous tubule destruction, testicular edema, degeneration of Leydig cells, and prostate acini damage. All together affect the epididymal semen quality and quantity including sperm viability, motility, and count. Interestingly, ChV could restore the testicular architecture and spermatozoa regeneration accompanied by semen quality improvement and increased reproductive hormones including testosterone. On the other side, ChV suppresses reactive oxygen species (ROS) formation via enhancement the antioxidant-related genes in the testicular tissue including SOD, CAT, GSH, and MDA and maintaining spermatocyte survival via suppression of apoptotic related genes including caspase3 and activating steroidogenic related genes including StAR and HSD17β3 in the cadmium-treated testes. In this study, ChV could enhance male fertility under normal or stressful conditions and ameliorate the adverse effects of hazardous heavy metals that are widely distributed in our environment.
Collapse
Affiliation(s)
- Mayada R Farag
- Forensic Medicine and Toxicology Department, Veterinary Medicine Faculty, Zagazig University, Zagazig, 44519, Egypt.
| | - Nesma I El-Naseery
- Department of Histology and Cytology, Faculty of Veterinary Medicine, Zagazig University, Zagazig, 44519, Egypt
| | - Eman I El Behery
- Anatomy and Embryology Department, Veterinary Medicine Faculty, Zagazig University, Zagazig, 44519, Egypt
| | - Doaa S Nouh
- Anatomy and Embryology Department, Veterinary Medicine Faculty, Zagazig University, Zagazig, 44519, Egypt
| | - Amany El-Mleeh
- Department of Pharmacology, Faculty of Veterinary Medicine, Menoufia University, Shebin Elkoum, 32511, Egypt
| | - Ismail M A Mostafa
- Biochemistry and Molecular Biology Department, Faculty of Veterinary Medicine, Cairo University, Giza, 12211, Egypt
| | - Mahmoud Alagawany
- Poultry Department, Faculty of Agriculture, Zagazig University, Zagazig, 44519, Egypt
| | - Alessandro Di Cerbo
- School of Biosciences and Veterinary Medicine, University of Camerino, Matelica, Italy
| | - Mahmoud M Azzam
- Department of Animal Production College of Food & Agriculture Sciences, King Saud University, Riyadh, 11451, Saudi Arabia
| | - Suzan A Mawed
- Zoology Department, Faculty of Science, Zagazig University, Zagazig, 44519, Egypt.
| |
Collapse
|
16
|
Parsons BL, Beal MA, Dearfield KL, Douglas GR, Gi M, Gollapudi BB, Heflich RH, Horibata K, Kenyon M, Long AS, Lovell DP, Lynch AM, Myers MB, Pfuhler S, Vespa A, Zeller A, Johnson GE, White PA. Severity of effect considerations regarding the use of mutation as a toxicological endpoint for risk assessment: A report from the 8th International Workshop on Genotoxicity Testing (IWGT). ENVIRONMENTAL AND MOLECULAR MUTAGENESIS 2024. [PMID: 38828778 DOI: 10.1002/em.22599] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Revised: 03/13/2024] [Accepted: 04/15/2024] [Indexed: 06/05/2024]
Abstract
Exposure levels without appreciable human health risk may be determined by dividing a point of departure on a dose-response curve (e.g., benchmark dose) by a composite adjustment factor (AF). An "effect severity" AF (ESAF) is employed in some regulatory contexts. An ESAF of 10 may be incorporated in the derivation of a health-based guidance value (HBGV) when a "severe" toxicological endpoint, such as teratogenicity, irreversible reproductive effects, neurotoxicity, or cancer was observed in the reference study. Although mutation data have been used historically for hazard identification, this endpoint is suitable for quantitative dose-response modeling and risk assessment. As part of the 8th International Workshops on Genotoxicity Testing, a sub-group of the Quantitative Analysis Work Group (WG) explored how the concept of effect severity could be applied to mutation. To approach this question, the WG reviewed the prevailing regulatory guidance on how an ESAF is incorporated into risk assessments, evaluated current knowledge of associations between germline or somatic mutation and severe disease risk, and mined available data on the fraction of human germline mutations expected to cause severe disease. Based on this review and given that mutations are irreversible and some cause severe human disease, in regulatory settings where an ESAF is used, a majority of the WG recommends applying an ESAF value between 2 and 10 when deriving a HBGV from mutation data. This recommendation may need to be revisited in the future if direct measurement of disease-causing mutations by error-corrected next generation sequencing clarifies selection of ESAF values.
Collapse
Affiliation(s)
- Barbara L Parsons
- Division of Genetic and Molecular Toxicology, National Center for Toxicological Research, U.S. Food and Drug Administration, Jefferson, Arkansas, USA
| | - Marc A Beal
- Bureau of Chemical Safety, Health Products and Food Branch, Health Canada, Ottawa, Ontario, Canada
| | - Kerry L Dearfield
- U.S. Environmental Protection Agency and U.S. Department of Agriculture, Washington, DC, USA
| | - George R Douglas
- Environmental Health Science and Research Bureau, Healthy Environments and Consumer Safety Branch, Health Canada, Ottawa, Ontario, Canada
| | - Min Gi
- Department of Environmental Risk Assessment, Osaka Metropolitan University Graduate School of Medicine, Osaka, Japan
| | | | - Robert H Heflich
- Division of Genetic and Molecular Toxicology, National Center for Toxicological Research, U.S. Food and Drug Administration, Jefferson, Arkansas, USA
| | | | - Michelle Kenyon
- Portfolio and Regulatory Strategy, Drug Safety Research and Development, Pfizer, Groton, Connecticut, USA
| | - Alexandra S Long
- Existing Substances Risk Assessment Bureau, Healthy Environments and Consumer Safety Branch, Health Canada, Ottawa, Ontario, Canada
| | - David P Lovell
- Population Health Research Institute, St George's Medical School, University of London, London, UK
| | | | - Meagan B Myers
- Division of Genetic and Molecular Toxicology, National Center for Toxicological Research, U.S. Food and Drug Administration, Jefferson, Arkansas, USA
| | | | - Alisa Vespa
- Pharmaceutical Drugs Directorate, Health Products and Food Branch, Health Canada, Ottawa, Ontario, Canada
| | - Andreas Zeller
- Pharmaceutical Sciences, pRED Innovation Center Basel, Hoffmann-La Roche Ltd, Basel, Switzerland
| | - George E Johnson
- Swansea University Medical School, Swansea University, Swansea, Wales, UK
| | - Paul A White
- Environmental Health Science and Research Bureau, Healthy Environments and Consumer Safety Branch, Health Canada, Ottawa, Ontario, Canada
| |
Collapse
|
17
|
Pham AH, Emori C, Ishikawa-Yamauchi Y, Tokuhiro K, Kamoshita M, Fujihara Y, Ikawa M. Thirteen Ovary-Enriched Genes Are Individually Not Essential for Female Fertility in Mice. Cells 2024; 13:802. [PMID: 38786026 PMCID: PMC11119756 DOI: 10.3390/cells13100802] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Revised: 05/01/2024] [Accepted: 05/03/2024] [Indexed: 05/25/2024] Open
Abstract
Infertility is considered a global health issue as it currently affects one in every six couples, with female factors reckoned to contribute to partly or solely 50% of all infertility cases. Over a thousand genes are predicted to be highly expressed in the female reproductive system and around 150 genes in the ovary. However, some of their functions in fertility remain to be elucidated. In this study, 13 ovary and/or oocyte-enriched genes (Ccdc58, D930020B18Rik, Elobl, Fbxw15, Oas1h, Nlrp2, Pramel34, Pramel47, Pkd1l2, Sting1, Tspan4, Tubal3, Zar1l) were individually knocked out by the CRISPR/Cas9 system. Mating tests showed that these 13 mutant mouse lines were capable of producing offspring. In addition, we observed the histology section of ovaries and performed in vitro fertilization in five mutant mouse lines. We found no significant anomalies in terms of ovarian development and fertilization ability. In this study, 13 different mutant mouse lines generated by CRISPR/Cas9 genome editing technology revealed that these 13 genes are individually not essential for female fertility in mice.
Collapse
Affiliation(s)
- Anh Hoang Pham
- Research Institute for Microbial Diseases, Osaka University, Osaka 565-0871, Japan; (A.H.P.); (C.E.); (M.K.); (Y.F.)
- Graduate School of Pharmaceutical Sciences, Osaka University, Osaka 565-0871, Japan
| | - Chihiro Emori
- Research Institute for Microbial Diseases, Osaka University, Osaka 565-0871, Japan; (A.H.P.); (C.E.); (M.K.); (Y.F.)
| | - Yu Ishikawa-Yamauchi
- Department of Regenerative Medicine, Yokohama City University Graduate School of Medicine, Yokohama 236-0027, Japan;
- The Institute of Medical Science, The University of Tokyo, Tokyo 108-8639, Japan
| | - Keizo Tokuhiro
- Department of Genome Editing, Institute of Biomedical Science, Kansai Medical University, Osaka 573-1191, Japan;
| | - Maki Kamoshita
- Research Institute for Microbial Diseases, Osaka University, Osaka 565-0871, Japan; (A.H.P.); (C.E.); (M.K.); (Y.F.)
| | - Yoshitaka Fujihara
- Research Institute for Microbial Diseases, Osaka University, Osaka 565-0871, Japan; (A.H.P.); (C.E.); (M.K.); (Y.F.)
- Department of Advanced Medical Technologies, National Cerebral and Cardiovascular Center, Osaka 564-8565, Japan
| | - Masahito Ikawa
- Research Institute for Microbial Diseases, Osaka University, Osaka 565-0871, Japan; (A.H.P.); (C.E.); (M.K.); (Y.F.)
- Graduate School of Pharmaceutical Sciences, Osaka University, Osaka 565-0871, Japan
- The Institute of Medical Science, The University of Tokyo, Tokyo 108-8639, Japan
- Center for Infectious Disease Education and Research, Osaka University, Osaka 565-0871, Japan
| |
Collapse
|
18
|
Aitken RJ. What is driving the global decline of human fertility? Need for a multidisciplinary approach to the underlying mechanisms. FRONTIERS IN REPRODUCTIVE HEALTH 2024; 6:1364352. [PMID: 38726051 PMCID: PMC11079147 DOI: 10.3389/frph.2024.1364352] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Accepted: 04/03/2024] [Indexed: 05/12/2024] Open
Abstract
An intense period of human population expansion over the past 250 years is about to cease. Total fertility rates are falling dramatically all over the world such that highly industrialized nations, including China and the tiger economies of SE Asia, will see their populations decline significantly in the coming decades. The socioeconomic, geopolitical and environmental ramifications of this change are considerable and invite a multidisciplinary consideration of the underlying mechanisms. In the short-term, socioeconomic factors, particularly urbanization and delayed childbearing are powerful drivers of reduced fertility. In parallel, lifestyle factors such as obesity and the presence of numerous reproductive toxicants in the environment, including air-borne pollutants, nanoplastics and electromagnetic radiation, are seriously compromising reproductive health. In the longer term, it is hypothesized that the reduction in family size that accompanies the demographic transition will decrease selection pressure on high fertility genes leading to a progressive loss of human fecundity. Paradoxically, the uptake of assisted reproductive technologies at scale, may also contribute to such fecundity loss by encouraging the retention of poor fertility genotypes within the population. Since the decline in fertility rate that accompanies the demographic transition appears to be ubiquitous, the public health implications for our species are potentially devastating.
Collapse
Affiliation(s)
- Robert John Aitken
- Priority Research Centre for Reproductive Science, Discipline of Biological Sciences, School of Environmental and Life Sciences, College of Engineering Science and Environment, University of Newcastle, Callaghan, NSW, Australia
- Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
| |
Collapse
|
19
|
Olszewska M, Malcher A, Stokowy T, Pollock N, Berman AJ, Budkiewicz S, Kamieniczna M, Jackowiak H, Suszynska-Zajczyk J, Jedrzejczak P, Yatsenko AN, Kurpisz M. Effects of Tcte1 knockout on energy chain transportation and spermatogenesis: implications for male infertility. Hum Reprod Open 2024; 2024:hoae020. [PMID: 38650655 PMCID: PMC11035007 DOI: 10.1093/hropen/hoae020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Revised: 03/08/2024] [Indexed: 04/25/2024] Open
Abstract
STUDY QUESTION Is the Tcte1 mutation causative for male infertility? SUMMARY ANSWER Our collected data underline the complex and devastating effect of the single-gene mutation on the testicular molecular network, leading to male reproductive failure. WHAT IS KNOWN ALREADY Recent data have revealed mutations in genes related to axonemal dynein arms as causative for morphology and motility abnormalities in spermatozoa of infertile males, including dysplasia of fibrous sheath (DFS) and multiple morphological abnormalities in the sperm flagella (MMAF). The nexin-dynein regulatory complex (N-DRC) coordinates the dynein arm activity and is built from the DRC1-DRC7 proteins. DRC5 (TCTE1), one of the N-DRC elements, has already been reported as a candidate for abnormal sperm flagella beating; however, only in a restricted manner with no clear explanation of respective observations. STUDY DESIGN SIZE DURATION Using the CRISPR/Cas9 genome editing technique, a mouse Tcte1 gene knockout line was created on the basis of the C57Bl/6J strain. The mouse reproductive potential, semen characteristics, testicular gene expression levels, sperm ATP, and testis apoptosis level measurements were then assessed, followed by visualization of N-DRC proteins in sperm, and protein modeling in silico. Also, a pilot genomic sequencing study of samples from human infertile males (n = 248) was applied for screening of TCTE1 variants. PARTICIPANTS/MATERIALS SETTING METHODS To check the reproductive potential of KO mice, adult animals were crossed for delivery of three litters per caged pair, but for no longer than for 6 months, in various combinations of zygosity. All experiments were performed for wild-type (WT, control group), heterozygous Tcte1+/- and homozygous Tcte1-/- male mice. Gross anatomy was performed on testis and epididymis samples, followed by semen analysis. Sequencing of RNA (RNAseq; Illumina) was done for mice testis tissues. STRING interactions were checked for protein-protein interactions, based on changed expression levels of corresponding genes identified in the mouse testis RNAseq experiments. Immunofluorescence in situ staining was performed to detect the N-DRC complex proteins: Tcte1 (Drc5), Drc7, Fbxl13 (Drc6), and Eps8l1 (Drc3) in mouse spermatozoa. To determine the amount of ATP in spermatozoa, the luminescence level was measured. In addition, immunofluorescence in situ staining was performed to check the level of apoptosis via caspase 3 visualization on mouse testis samples. DNA from whole blood samples of infertile males (n = 137 with non-obstructive azoospermia or cryptozoospermia, n = 111 samples with a spectrum of oligoasthenoteratozoospermia, including n = 47 with asthenozoospermia) was extracted to perform genomic sequencing (WGS, WES, or Sanger). Protein prediction modeling of human-identified variants and the exon 3 structure deleted in the mouse knockout was also performed. MAIN RESULTS AND THE ROLE OF CHANCE No progeny at all was found for the homozygous males which were revealed to have oligoasthenoteratozoospermia, while heterozygous animals were fertile but manifested oligozoospermia, suggesting haploinsufficiency. RNA-sequencing of the testicular tissue showed the influence of Tcte1 mutations on the expression pattern of 21 genes responsible for mitochondrial ATP processing or linked with apoptosis or spermatogenesis. In Tcte1-/- males, the protein was revealed in only residual amounts in the sperm head nucleus and was not transported to the sperm flagella, as were other N-DRC components. Decreased ATP levels (2.4-fold lower) were found in the spermatozoa of homozygous mice, together with disturbed tail:midpiece ratios, leading to abnormal sperm tail beating. Casp3-positive signals (indicating apoptosis) were observed in spermatogonia only, at a similar level in all three mouse genotypes. Mutation screening of human infertile males revealed one novel and five ultra-rare heterogeneous variants (predicted as disease-causing) in 6.05% of the patients studied. Protein prediction modeling of identified variants revealed changes in the protein surface charge potential, leading to disruption in helix flexibility or its dynamics, thus suggesting disrupted interactions of TCTE1 with its binding partners located within the axoneme. LARGE SCALE DATA All data generated or analyzed during this study are included in this published article and its supplementary information files. RNAseq data are available in the GEO database (https://www.ncbi.nlm.nih.gov/geo/) under the accession number GSE207805. The results described in the publication are based on whole-genome or exome sequencing data which includes sensitive information in the form of patient-specific germline variants. Information regarding such variants must not be shared publicly following European Union legislation, therefore access to raw data that support the findings of this study are available from the corresponding author upon reasonable request. LIMITATIONS REASONS FOR CAUTION In the study, the in vitro fertilization performance of sperm from homozygous male mice was not checked. WIDER IMPLICATIONS OF THE FINDINGS This study contains novel and comprehensive data concerning the role of TCTE1 in male infertility. The TCTE1 gene is the next one that should be added to the 'male infertility list' because of its crucial role in spermatogenesis and proper sperm functioning. STUDY FUNDING/COMPETING INTERESTS This work was supported by National Science Centre in Poland, grants no.: 2015/17/B/NZ2/01157 and 2020/37/B/NZ5/00549 (to M.K.), 2017/26/D/NZ5/00789 (to A.M.), and HD096723, GM127569-03, NIH SAP #4100085736 PA DoH (to A.N.Y.). The authors declare that there is no conflict of interest that could be perceived as prejudicing the impartiality of the research reported.
Collapse
Affiliation(s)
- Marta Olszewska
- Institute of Human Genetics, Polish Academy of Sciences, Poznan, Poland
| | - Agnieszka Malcher
- Institute of Human Genetics, Polish Academy of Sciences, Poznan, Poland
| | - Tomasz Stokowy
- Scientific Computing Group, IT Division, University of Bergen, Bergen, Norway
| | - Nijole Pollock
- Department of OB/GYN and Reproductive Sciences, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Andrea J Berman
- Department of Biological Sciences, University of Pittsburgh, Pittsburgh, PA, USA
| | - Sylwia Budkiewicz
- Institute of Human Genetics, Polish Academy of Sciences, Poznan, Poland
| | | | - Hanna Jackowiak
- Department of Histology and Embryology, Poznan University of Life Sciences, Poznan, Poland
| | | | - Piotr Jedrzejczak
- Division of Infertility and Reproductive Endocrinology, Department of Gynecology, Obstetrics and Gynecological Oncology, Poznan University of Medical Sciences, Poznan, Poland
| | - Alexander N Yatsenko
- Department of OB/GYN and Reproductive Sciences, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Maciej Kurpisz
- Institute of Human Genetics, Polish Academy of Sciences, Poznan, Poland
| |
Collapse
|
20
|
Zhang JL, Xu MF, Chen J, Wei YL, She ZY. Kinesin-7 CENP-E mediates chromosome alignment and spindle assembly checkpoint in meiosis I. Chromosoma 2024; 133:149-168. [PMID: 38456964 DOI: 10.1007/s00412-024-00818-w] [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: 03/27/2023] [Revised: 02/05/2024] [Accepted: 02/26/2024] [Indexed: 03/09/2024]
Abstract
In eukaryotes, meiosis is the genetic basis for sexual reproduction, which is important for chromosome stability and species evolution. The defects in meiosis usually lead to chromosome aneuploidy, reduced gamete number, and genetic diseases, but the pathogenic mechanisms are not well clarified. Kinesin-7 CENP-E is a key regulator in chromosome alignment and spindle assembly checkpoint in cell division. However, the functions and mechanisms of CENP-E in male meiosis remain largely unknown. In this study, we have revealed that the CENP-E gene was highly expressed in the rat testis. CENP-E inhibition influences chromosome alignment and spindle organization in metaphase I spermatocytes. We have found that a portion of misaligned homologous chromosomes is located at the spindle poles after CENP-E inhibition, which further activates the spindle assembly checkpoint during the metaphase-to-anaphase transition in rat spermatocytes. Furthermore, CENP-E depletion leads to abnormal spermatogenesis, reduced sperm count, and abnormal sperm head structure. Our findings have elucidated that CENP-E is essential for homologous chromosome alignment and spindle assembly checkpoint in spermatocytes, which further contribute to chromosome stability and sperm cell quality during spermatogenesis.
Collapse
Affiliation(s)
- Jing-Lian Zhang
- Department of Cell Biology and Genetics, The School of Basic Medical Sciences, Fujian Medical University, Fuzhou, 350122, Fujian, China
- Key Laboratory of Stem Cell Engineering and Regenerative Medicine, Fujian Province University, Fuzhou, 350122, Fujian, China
| | - Meng-Fei Xu
- Department of Cell Biology and Genetics, The School of Basic Medical Sciences, Fujian Medical University, Fuzhou, 350122, Fujian, China
- Key Laboratory of Stem Cell Engineering and Regenerative Medicine, Fujian Province University, Fuzhou, 350122, Fujian, China
| | - Jie Chen
- Department of Cell Biology and Genetics, The School of Basic Medical Sciences, Fujian Medical University, Fuzhou, 350122, Fujian, China
- Key Laboratory of Stem Cell Engineering and Regenerative Medicine, Fujian Province University, Fuzhou, 350122, Fujian, China
| | - Ya-Lan Wei
- Medical Research Center, Fujian Maternity and Child Health Hospital, Fuzhou, 350001, Fujian, China
- College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, Fuzhou, 350122, Fujian, China
| | - Zhen-Yu She
- Department of Cell Biology and Genetics, The School of Basic Medical Sciences, Fujian Medical University, Fuzhou, 350122, Fujian, China.
- Key Laboratory of Stem Cell Engineering and Regenerative Medicine, Fujian Province University, Fuzhou, 350122, Fujian, China.
| |
Collapse
|
21
|
Abdelnour SA, Khalil WA, Khalifa NE, Khalil FMA, Hassan MAE. L-Proline: A Promising Tool for Boosting Cryotolerance and Fertilizing Ability of Cryopreserved Sperm in Animals. Anim Reprod Sci 2024; 263:107429. [PMID: 38382197 DOI: 10.1016/j.anireprosci.2024.107429] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Revised: 01/11/2024] [Accepted: 02/06/2024] [Indexed: 02/23/2024]
Abstract
Sperm cryopreservation technology significantly contributes to the safeguarding of genetic resources, particularly for endangered species, and supports the use of artificial insemination in domestic animals. Therefore, cryopreservation can negatively affect sperm health and function leading to reduce the freezing ability and fertility potential. Therefore, it is essential to prioritize the improvement of cryotolerance in cryopreserved sperm to enhance reproductive efficiency and ensure sustainability in livestock herds. The main reason for sperm dysfunction after thawing may be related to the excessive amount of oxidative stress (OS) produced during cryopreservation. Scientists have different ways for counteracting this OS including the use of plant extracts, enzymes, minerals, anti-freezing proteins, and amino acids. Recently, one such amino acid is L-proline (LP), which has multiple roles such as osmotic and OS defense, nitrogen, and carbon metabolism, as well as cell survival and signaling. LP has been found in seminal plasma and has recently been added to the freezing extender to improve the various post-thaw parameters of sperm. This improvement is related to the ability of LP to reduce the OS, sustain the plasma membrane and to act as an osmoregulatory agent. Moreover, LP can suppress cell apoptosis by modulating intracellular redox in sperm. This review addresses the ongoing research on the addition of L-proline as an osmoregulatory agent in freezing extenders to increase the cryotolerance of animal spermatozoa to freeze-thaw.
Collapse
Affiliation(s)
- Sameh A Abdelnour
- Department of Animal Production, Faculty of Agriculture, Zagazig University, Zagazig, Egypt.
| | - Wael A Khalil
- Department of Animal Production, Faculty of Agriculture, Mansoura University Mansoura 35516, Egypt.
| | - Norhan E Khalifa
- Department of Physiology, Faculty of Veterinary Medicine, Matrouh University, Fuka, Matrouh 51744, Egypt
| | - Fatma Mohamed Ameen Khalil
- Department of Biology, College of Science and Arts, King Khalid University, Mohayil Asir Abha 61421, Saudi Arabia
| | - Mahmoud A E Hassan
- Animal Production Research Institute, Agriculture Research Centre, Ministry of Agriculture, Dokki, Giza 12619, Egypt
| |
Collapse
|
22
|
Zheng H, Gong C, Li J, Hou J, Gong X, Zhu X, Deng H, Wu H, Zhang F, Shi Q, Zhou J, Shi B, Yang X, Xi Y. CCDC157 is essential for sperm differentiation and shows oligoasthenoteratozoospermia-related mutations in men. J Cell Mol Med 2024; 28:e18215. [PMID: 38509755 PMCID: PMC10955179 DOI: 10.1111/jcmm.18215] [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: 10/30/2023] [Revised: 01/09/2024] [Accepted: 02/09/2024] [Indexed: 03/22/2024] Open
Abstract
Oligoasthenoteratospermia (OAT), characterized by abnormally low sperm count, poor sperm motility, and abnormally high number of deformed spermatozoa, is an important cause of male infertility. Its genetic basis in many affected individuals remains unknown. Here, we found that CCDC157 variants are associated with OAT. In two cohorts, a 21-bp (g.30768132_30768152del21) and/or 24-bp (g.30772543_30772566del24) deletion of CCDC157 were identified in five sporadic OAT patients, and 2 cases within one pedigree. In a mouse model, loss of Ccdc157 led to male sterility with OAT-like phenotypes. Electron microscopy revealed misstructured acrosome and abnormal head-tail coupling apparatus in the sperm of Ccdc157-null mice. Comparative transcriptome analysis showed that the Ccdc157 mutation alters the expressions of genes involved in cell migration/motility and Golgi components. Abnormal Golgi apparatus and decreased expressions of genes involved in acrosome formation and lipid metabolism were detected in Ccdc157-deprived mouse germ cells. Interestingly, we attempted to treat infertile patients and Ccdc157 mutant mice with a Chinese medicine, Huangjin Zanyu, which improved the fertility in one patient and most mice that carried the heterozygous mutation in CCDC157. Healthy offspring were produced. Our study reveals CCDC157 is essential for sperm maturation and may serve as a marker for diagnosis of OAT.
Collapse
Affiliation(s)
- Huimei Zheng
- Division of Human Reproduction and Developmental Genetics, the Women's HospitalZhejiang University School of MedicineHangzhouChina
| | - Chenjia Gong
- Hefei National Laboratory for Physical Sciences at Microscale, the First Affiliated Hospital of USTC, USTC‐SJH Joint Center for Human Reproduction and Genetics, The CAS Key Laboratory of Innate Immunity and Chronic Diseases, School of Life Sciences, CAS Center for Excellence in Molecular Cell Science, Collaborative Innovation Center of Genetics and DevelopmentUniversity of Science and Technology of ChinaHefeiChina
| | - Jingping Li
- Division of Human Reproduction and Developmental Genetics, the Women's HospitalZhejiang University School of MedicineHangzhouChina
| | - Jiaru Hou
- Division of Human Reproduction and Developmental Genetics, the Women's HospitalZhejiang University School of MedicineHangzhouChina
- Institute of GeneticsZhejiang UniversityYiwuChina
- Center for Genetic Medicine, the Fourth Affiliated HospitalZhejiang University School of MedicineYiwuChina
| | - Xinhan Gong
- Division of Human Reproduction and Developmental Genetics, the Women's HospitalZhejiang University School of MedicineHangzhouChina
- Institute of GeneticsZhejiang UniversityYiwuChina
- Center for Genetic Medicine, the Fourth Affiliated HospitalZhejiang University School of MedicineYiwuChina
| | - Xinhai Zhu
- College of Life SciencesZhejiang UniversityHangzhouChina
| | - Huan Deng
- Division of Human Reproduction and Developmental Genetics, the Women's HospitalZhejiang University School of MedicineHangzhouChina
- Institute of GeneticsZhejiang UniversityYiwuChina
- Center for Genetic Medicine, the Fourth Affiliated HospitalZhejiang University School of MedicineYiwuChina
| | - Haoyue Wu
- Division of Human Reproduction and Developmental Genetics, the Women's HospitalZhejiang University School of MedicineHangzhouChina
- Institute of GeneticsZhejiang UniversityYiwuChina
- Center for Genetic Medicine, the Fourth Affiliated HospitalZhejiang University School of MedicineYiwuChina
| | - Fengbin Zhang
- Division of Human Reproduction and Developmental Genetics, the Women's HospitalZhejiang University School of MedicineHangzhouChina
| | - Qinghua Shi
- Hefei National Laboratory for Physical Sciences at Microscale, the First Affiliated Hospital of USTC, USTC‐SJH Joint Center for Human Reproduction and Genetics, The CAS Key Laboratory of Innate Immunity and Chronic Diseases, School of Life Sciences, CAS Center for Excellence in Molecular Cell Science, Collaborative Innovation Center of Genetics and DevelopmentUniversity of Science and Technology of ChinaHefeiChina
| | - Jianteng Zhou
- Hefei National Laboratory for Physical Sciences at Microscale, the First Affiliated Hospital of USTC, USTC‐SJH Joint Center for Human Reproduction and Genetics, The CAS Key Laboratory of Innate Immunity and Chronic Diseases, School of Life Sciences, CAS Center for Excellence in Molecular Cell Science, Collaborative Innovation Center of Genetics and DevelopmentUniversity of Science and Technology of ChinaHefeiChina
| | - Baolu Shi
- Hefei National Laboratory for Physical Sciences at Microscale, the First Affiliated Hospital of USTC, USTC‐SJH Joint Center for Human Reproduction and Genetics, The CAS Key Laboratory of Innate Immunity and Chronic Diseases, School of Life Sciences, CAS Center for Excellence in Molecular Cell Science, Collaborative Innovation Center of Genetics and DevelopmentUniversity of Science and Technology of ChinaHefeiChina
| | - Xiaohang Yang
- Division of Human Reproduction and Developmental Genetics, the Women's HospitalZhejiang University School of MedicineHangzhouChina
- Institute of GeneticsZhejiang UniversityYiwuChina
- Center for Genetic Medicine, the Fourth Affiliated HospitalZhejiang University School of MedicineYiwuChina
| | - Yongmei Xi
- Division of Human Reproduction and Developmental Genetics, the Women's HospitalZhejiang University School of MedicineHangzhouChina
- Institute of GeneticsZhejiang UniversityYiwuChina
- Center for Genetic Medicine, the Fourth Affiliated HospitalZhejiang University School of MedicineYiwuChina
| |
Collapse
|
23
|
Bhattacharya I, Sharma SS, Majumdar SS. Etiology of Male Infertility: an Update. Reprod Sci 2024; 31:942-965. [PMID: 38036863 DOI: 10.1007/s43032-023-01401-x] [Citation(s) in RCA: 27] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Accepted: 10/30/2023] [Indexed: 12/02/2023]
Abstract
Spermatogenesis is a complex process of germ cell division and differentiation that involves extensive cross-talk between the developing germ cells and the somatic testicular cells. Defective endocrine signaling and/or intrinsic defects within the testes can adversely affect spermatogenic progression, leading to subfertility/infertility. In recent years, male infertility has been recognized as a global public health concern, and research over the last few decades has elucidated the complex etiology of male infertility. Congenital reproductive abnormalities, genetic mutations, and endocrine/metabolic dysfunction have been demonstrated to be involved in infertility/subfertility in males. Furthermore, acquired factors like exposure to environmental toxicants and lifestyle-related disorders such as illicit use of psychoactive drugs have been shown to adversely affect spermatogenesis. Despite the large body of available scientific literature on the etiology of male infertility, a substantial proportion of infertility cases are idiopathic in nature, with no known cause. The inability to treat such idiopathic cases stems from poor knowledge about the complex regulation of spermatogenesis. Emerging scientific evidence indicates that defective functioning of testicular Sertoli cells (Sc) may be an underlying cause of infertility/subfertility in males. Sc plays an indispensable role in regulating spermatogenesis, and impaired functional maturation of Sc has been shown to affect fertility in animal models as well as humans, suggesting abnormal Sc as a potential underlying cause of reproductive insufficiency/failure in such cases of unexplained infertility. This review summarizes the major causes of infertility/subfertility in males, with an emphasis on infertility due to dysregulated Sc function.
Collapse
Affiliation(s)
- Indrashis Bhattacharya
- Department of Zoology, Central University of Kerala, Periye Campus, Kasaragod, 671320, Kerala, India.
| | - Souvik Sen Sharma
- National Institute of Animal Biotechnology, Hyderabad, 500 032, Telangana, India
| | - Subeer S Majumdar
- National Institute of Animal Biotechnology, Hyderabad, 500 032, Telangana, India.
- Gujarat Biotechnology University, Gandhinagar, GIFT City, Gandhinagar, 382355, Gujarat, India.
| |
Collapse
|
24
|
Long C, Benny P, Yap J, Lee J, Huang Z. A Systematic Review of Genetics and Reproductive Health Outcomes: Asian Perspective. Reprod Sci 2024; 31:309-319. [PMID: 37524971 DOI: 10.1007/s43032-023-01311-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Accepted: 07/14/2023] [Indexed: 08/02/2023]
Abstract
In the last four decades, advances in assisted reproductive technology (ART) have offered hope to individuals with fertility problems to conceive. However, a closer examination of the clinical outcomes of ART shows a stark contrast in Asian women compared to Caucasians, with majority of studies reporting lower reproductive success among Asian women. We performed a systematic review to elucidate the genes associated with ART clinical outcomes, with a focus on Asian ethnicities. We completed a database search to identify all studies associated with reproductive outcomes in women of different ethnic backgrounds. Following PRISMA, 128 studies were analyzed. Pathway analysis of gene sets was done using Cytoscapev3.4.0. We observed that age at menarche (AAM) was correlated with the timing of the first pregnancy, with Hawaiians having the lowest age (22.2 years) and Japanese the highest age (25.0 years). LIN28 mutations were associated with AAM and prevalent in both Chinese and American populations. FMR1 was most associated with ovarian reserve. Network analysis highlighted a close association between FMR1, FSHR, ESR1, BMP15, and INHA, through biological functions affecting menstrual cycle and hypothalamic-pituitary axis and therefore ovarian follicle development. Leveraging these findings, we propose the development of a personalized, ethnic-specific biomarker panel which would enhance patient stratification to address every woman's unique reproductive potential.
Collapse
Affiliation(s)
- Cheryl Long
- Department of Obstetrics and Gynaecology, National University Hospital, National University of Singapore, 1E Kent Ridge Rd, Level 12 NUHS Tower Block, Singapore, 119228, Singapore
| | - Paula Benny
- Department of Obstetrics and Gynaecology, National University Hospital, National University of Singapore, 1E Kent Ridge Rd, Level 12 NUHS Tower Block, Singapore, 119228, Singapore
- NUS Bia-Echo Asia Centre of Reproductive Longevity and Equality, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Jeannie Yap
- Department of Obstetrics and Gynaecology, National University Hospital, National University of Singapore, 1E Kent Ridge Rd, Level 12 NUHS Tower Block, Singapore, 119228, Singapore
| | - Jovin Lee
- Department of Obstetrics and Gynaecology, National University Hospital, National University of Singapore, 1E Kent Ridge Rd, Level 12 NUHS Tower Block, Singapore, 119228, Singapore
- NUS Bia-Echo Asia Centre of Reproductive Longevity and Equality, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Zhongwei Huang
- Department of Obstetrics and Gynaecology, National University Hospital, National University of Singapore, 1E Kent Ridge Rd, Level 12 NUHS Tower Block, Singapore, 119228, Singapore.
- NUS Bia-Echo Asia Centre of Reproductive Longevity and Equality, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.
| |
Collapse
|
25
|
Salmanov AG, Yuzko OM, Tofan BY, Artyomenko VV, Korniyenko SM, Rud VO, Dyndar OA, Kovalyshyn OA, Nykoniuk TR, Nastradina NM. Factors associated with female infertility in Ukraine: results a multicenter study. WIADOMOSCI LEKARSKIE (WARSAW, POLAND : 1960) 2024; 77:790-799. [PMID: 38865639 DOI: 10.36740/wlek202404127] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2024]
Abstract
OBJECTIVE Aim: To determine the current prevalence of female infertility and characterize and identify risk factors associated with infertility in Ukraine. PATIENTS AND METHODS Materials and Methods: Multicenter prospective cohort study was conducted from January 2021 to December 2023 in twelve medical centers from nine regions of Ukraine. Definitions of infertility were adapted from the World Health Organization. According to the data collected from questionnaire, participants were divided into infertile and fertile groups and analyzed associated factors. RESULTS Results: Among all the 7,618 participants in this study, the prevalence of female infertility was 24.3%. The prevalence of primary infertility was 5.9%, and the prevalence of secondary infertility was 18.4%. In logistic multivariate regression analyses, female infertility was associated with age of women (p<0.001), age of first sexual intercourse (p<0.001), history of gynecological surgery (p<0.001), marital status (p<0.001), age of marriage (p<0.001), decreased ovarian reserve (DOR) (p=0.006), family history of infertility (p<0.001), history of cervicitis (p=0.007), history of surgical abortion (p<0.001), history of endometritis (p=0.027), bacterial vaginosis (p=0.023), and aerobic vaginitis (< 0.001). CONCLUSION Conclusions: Our data suggest a high prevalence of female infertility in Ukraine. The prevalence of secondary infertility was higher than primary infertility. Age of women, age of first sexual intercourse, history of gynecological surgery, marital status, age of marriage, DOR, family history of infertility, history of cervicitis, history of surgical abortion, history of endometritis, bacterial vaginosis, and aerobic vaginitis were associated with infertility.
Collapse
Affiliation(s)
- Aidyn G Salmanov
- SHUPYK NATIONAL HEALTHCARE UNIVERSITY OF UKRAINE, KYIV, UKRAINE; INSTITUTE OF PEDIATRICS, OBSTETRICS AND GYNECOLOGY OF THE NATIONAL ACADEMY OF MEDICAL SCIENCES OF UKRAINE, KYIV, UKRAINE
| | | | - Bohdan Yu Tofan
- SHUPYK NATIONAL HEALTHCARE UNIVERSITY OF UKRAINE, KYIV, UKRAINE
| | | | | | - Victor O Rud
- NATIONAL PIROGOV MEMORIAL MEDICAL UNIVERSITY, VINNYTSIA, UKRAINE
| | | | | | | | | |
Collapse
|
26
|
Lin D, Zou Y, Li X, Wang J, Xiao Q, Gao X, Lin F, Zhang N, Jiao M, Guo Y, Teng Z, Li S, Wei Y, Zhou F, Yin R, Zhang S, Xing L, Xu W, Wu X, Yang B, Xiao K, Wu C, Tao Y, Yang X, Zhang J, Hu S, Dong S, Li X, Ye S, Hong Z, Pan Y, Yang Y, Sun H, Cao G. MGA-seq: robust identification of extrachromosomal DNA and genetic variants using multiple genetic abnormality sequencing. Genome Biol 2023; 24:247. [PMID: 37904244 PMCID: PMC10614391 DOI: 10.1186/s13059-023-03081-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2023] [Accepted: 10/04/2023] [Indexed: 11/01/2023] Open
Abstract
Genomic abnormalities are strongly associated with cancer and infertility. In this study, we develop a simple and efficient method - multiple genetic abnormality sequencing (MGA-Seq) - to simultaneously detect structural variation, copy number variation, single-nucleotide polymorphism, homogeneously staining regions, and extrachromosomal DNA (ecDNA) from a single tube. MGA-Seq directly sequences proximity-ligated genomic fragments, yielding a dataset with concurrent genome three-dimensional and whole-genome sequencing information, enabling approximate localization of genomic structural variations and facilitating breakpoint identification. Additionally, by utilizing MGA-Seq, we map focal amplification and oncogene coamplification, thus facilitating the exploration of ecDNA's transcriptional regulatory function.
Collapse
Affiliation(s)
- Da Lin
- Precision Research Center for Refractory Diseases, Institute for Clinical Research, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Yanyan Zou
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, China
- College of Informatics, Huazhong Agricultural University, Wuhan, China
| | - Xinyu Li
- Precision Research Center for Refractory Diseases, Institute for Clinical Research, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jinyue Wang
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, China
- College of Bio-Medicine and Health, Huazhong Agricultural University, Wuhan, China
- College of Life Science and Technology, Huazhong Agricultural University, Wuhan, China
| | - Qin Xiao
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, China
- College of Bio-Medicine and Health, Huazhong Agricultural University, Wuhan, China
- College of Life Science and Technology, Huazhong Agricultural University, Wuhan, China
| | - Xiaochen Gao
- Precision Research Center for Refractory Diseases, Institute for Clinical Research, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Fei Lin
- Reproductive Medical Center, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Ningyuan Zhang
- Reproductive Medical Center, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Ming Jiao
- Department of Laboratory Animal Center, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yu Guo
- Department of Laboratory Animal Center, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhaowei Teng
- The First People's Hospital of Yunnan Province, Affiliated Hospital of Kunming University of Science and Technology, Kunming, China
| | - Shiyi Li
- Baylor College of Medicine, Houston, TX, USA
- Department of Radiation & Medical Oncology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Yongchang Wei
- Department of Radiation & Medical Oncology, Zhongnan Hospital of Wuhan University, Wuhan, China
- Hubei Key Laboratory of Tumor Biological Behaviors, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Fuling Zhou
- Department of Hematology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Rong Yin
- Department of Hematology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Siheng Zhang
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, China
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Lingyu Xing
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, China
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Weize Xu
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, China
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Xiaofeng Wu
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, China
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Bing Yang
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, China
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Ke Xiao
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, China
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Chengchao Wu
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, China
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Yingfeng Tao
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, China
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Xiaoqing Yang
- Hospital of Huazhong Agricultural University, Wuhan, China
| | - Jing Zhang
- Department of Medical Oncology, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Sheng Hu
- Department of Medical Oncology, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shuang Dong
- Department of Medical Oncology, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaoyu Li
- Department of Medical Oncology, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shengwei Ye
- Department of Gastrointestinal Surgery, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhidan Hong
- Dapartment of Reproductive Medicine Center, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Yihang Pan
- Precision Medicine Center, Scientific Research Center, School of Medicine, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, China
| | - Yuqin Yang
- Department of Laboratory Animal Center, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Haixiang Sun
- Reproductive Medical Center, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China.
| | - Gang Cao
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, China.
- College of Bio-Medicine and Health, Huazhong Agricultural University, Wuhan, China.
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China.
- Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China.
| |
Collapse
|
27
|
Doulgeraki T, Papageorgopoulou M, Iliodromiti S. The genetic background of female reproductive disorders: a systematic review. Curr Opin Obstet Gynecol 2023; 35:426-433. [PMID: 37266690 DOI: 10.1097/gco.0000000000000896] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/03/2023]
Abstract
PURPOSE OF REVIEW Reproductive function is the interplay between environmental factors and the genetic footprint of each individual. The development in genetic analysis has strengthened its role in the investigation of female reproductive disorders, potential treatment options and provision of personalized care. Despite the increasing requirement of genetic testing, the evidence of the gene-disease relationships (GDR) is limited. We performed a systematic review exploring the associations between the most frequent female reproductive endocrine disorders associated with subfertility [including polycystic ovaries syndrome (PCOS), premature ovarian failure (POI) and hypogonadotropic hypogonadism] and their genetic background in order to summarize current knowledge. METHODS A systematic review of relevant literature in accordance with PRISMA guidelines was conducted until July 2022. Data sources that were used are PubMed and Embase. RECENT FINDINGS A total of 55 studies were included from the 614 articles identified in the original search. We identified 384 genes associated with one or more of the included female reproductive disorders. The highest number of genes was found to be associated with POI ( N = 209), followed by hypogonadotropic hypogonadism ( N = 88) and PCOS ( N = 87). Four genes, including FSHR , LHβ , LEPR and SF1 were associated with multiple reproductive disorders implying common pathways in the development of those diseases. SUMMARY We provide an up-to-date summary of the currently known genes that are associated with three female reproductive disorders (PCOS, POI and hypogonadotropic hypogonadism). The role of genetic analysis in the field of impaired female reproduction may have a role in the diagnosis of female reproductive disorders and personalized patient care.
Collapse
Affiliation(s)
- Triada Doulgeraki
- Department of Obstetrics and Gynaecology, Royal London Hospital, Barts Health NHS Trust
| | - Maria Papageorgopoulou
- Women's Health Research Unit, Wolfson Institute of Population Health, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, United Kingdom
| | - Stamatina Iliodromiti
- Women's Health Research Unit, Wolfson Institute of Population Health, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, United Kingdom
| |
Collapse
|
28
|
Tang X, Ma J, Wang X, Long S, Wan L, Yu H, Yang J, Huang G, Lin T. A novel variant in CFAP69 causes asthenoteratozoospermia with treatable ART outcomes and a literature review. J Assist Reprod Genet 2023; 40:2175-2184. [PMID: 37392306 PMCID: PMC10440328 DOI: 10.1007/s10815-023-02873-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Accepted: 06/20/2023] [Indexed: 07/03/2023] Open
Abstract
PURPOSE Multiple morphological abnormalities of the sperm flagella (MMAF) are a severe form of sperm defect causing male infertility. Previous studies identified the variants in the CFAP69 gene as a MMAF-associated factor, but few cases have been reported. This study was performed to identify additional variants in CFAP69 and describe the semen characteristics and outcomes of assisted reproductive technology (ART) in CFAP69-affected couples. METHODS Genetic testing with next-generation sequencing (NGS) panel of 22 MMAF-associated genes and Sanger sequencing was performed in a cohort of 35 infertile males with MMAF to identify pathogenic variants. Morphological, ultrastructural, and immunostaining analyses were performed to investigate the characteristics of probands' spermatozoa. ART with intracytoplasmic sperm injection (ICSI) was carried out for the affected couples to get their own progenies. RESULTS We identified a novel frameshift variant in CFAP69 (c.2061dup, p. Pro688Thrfs*5) from a MMAF-affected infertile male with low sperm motility and malformed morphology of sperm. Furthermore, transmission electron microscopy and immunofluorescence staining revealed that the variant induced the aberrant ultrastructure and reduction of CFAP69 expression in the proband's spermatozoa. Moreover, the partner of the proband birthed a healthy girl through ICSI. CONCLUSIONS This study expanded the variant spectrum of CFAP69 and described the good outcome of ART treatment with ICSI, which is beneficial to the molecular diagnosis, genetic counseling, and treatment of infertile males with MMAF in the future.
Collapse
Affiliation(s)
- Xiangrong Tang
- Center for Reproductive Medicine, Women and Children's Hospital of Chongqing Medical University, Chongqing, 400013, China
- Chongqing Health Center for Women and Children, Chongqing, 400013, China
| | - Jing Ma
- Center for Reproductive Medicine, Women and Children's Hospital of Chongqing Medical University, Chongqing, 400013, China
- Chongqing Health Center for Women and Children, Chongqing, 400013, China
| | - Xinglin Wang
- Center for Reproductive Medicine, Women and Children's Hospital of Chongqing Medical University, Chongqing, 400013, China
- Chongqing Health Center for Women and Children, Chongqing, 400013, China
| | - Shunhua Long
- Center for Reproductive Medicine, Women and Children's Hospital of Chongqing Medical University, Chongqing, 400013, China
- Chongqing Health Center for Women and Children, Chongqing, 400013, China
| | - Ling Wan
- Chongqing Population and Family Planning Science and Technology Research Institute, Chongqing, 400020, China
| | - Haibing Yu
- Center for Reproductive Medicine, Women and Children's Hospital of Chongqing Medical University, Chongqing, 400013, China
- Chongqing Health Center for Women and Children, Chongqing, 400013, China
| | - Jigao Yang
- Chongqing Population and Family Planning Science and Technology Research Institute, Chongqing, 400020, China
| | - Guoning Huang
- Center for Reproductive Medicine, Women and Children's Hospital of Chongqing Medical University, Chongqing, 400013, China.
- Chongqing Health Center for Women and Children, Chongqing, 400013, China.
| | - Tingting Lin
- Center for Reproductive Medicine, Women and Children's Hospital of Chongqing Medical University, Chongqing, 400013, China.
- Chongqing Health Center for Women and Children, Chongqing, 400013, China.
| |
Collapse
|
29
|
Abdullah AA, Ahmed M, Oladokun A. Characterization and risk factors for unexplained female infertility in Sudan: A case-control study. World J Methodol 2023; 13:98-117. [PMID: 37456975 PMCID: PMC10348085 DOI: 10.5662/wjm.v13.i3.98] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/25/2022] [Revised: 02/08/2023] [Accepted: 03/15/2023] [Indexed: 06/20/2023] Open
Abstract
BACKGROUND Female infertility with unknown causes (unexplained) remains one of the mysteries in the reproductive health field, where the diagnostic evidence is still weak and the proposed treatments still work with unknown methods. However, several studies have proposed some possible causes and risk factors for unexplained female infertility. AIM To characterize and identify factors associated with unexplained infertility in Sudanese women. METHODS A matched (age and body mass index) case-control study was conducted from March 2021 to February 2022. The study samples were 210 women with unexplained infertility (UI) and 190 fertile women of reproductive age who were attending the maternity hospitals and fertility clinics in Khartoum, Sudan. The risk factors of unexplained infertility were identified using a structured, pre-tested questionnaire containing information on socio-demographic variables, anthropometrics, clinical diagnosis of infertility, behavioral factors, physical activity assessment, diversity, and consumption of different food groups by the study participants. RESULTS The results showed a higher proportion of women diagnosed with UI were residents of rural areas than controls (21.4% vs 11.1%, P < 0.05), and previous miscarriages and/or abortions were more common in fertile women compared with infertile women (13.16% vs 5.71%, P < 0.05). Additionally, infertile women had a significantly (P < 0.05) higher proportion of family history of infertility (explained and unexplained) compared with controls. Finally, after controlling for the effects of potentially confounding variables using multivariable logistic regression analysis, only marital status, family history of infertility, use of modern contraceptives, smoking, caffeine consumption, physical activity level, meals consumed, other vitamin-A-rich fruits and vegetables, and other vegetables were found to be significant (P < 0.05) factors associated with unexplained infertility among Sudanese women. CONCLUSION Married women with a family history of infertility who smoke and consume a high amount of caffeine, who live a sedentary lifestyle, and who consume more than two meals free of vitamin-A-rich fruits and/or vegetables and/or other vegetables per day are at the highest risk of developing unexplained infertility.
Collapse
Affiliation(s)
- Abdullah Abdulslam Abdullah
- Reproductive Health Sciences Program, Pan African University Life and Earth Sciences Institute, University of Ibadan, Ibadan 119, Oyo state, Nigeria
- Department of Obstetrics and Gynecology, College of Medicine, University of Ibadan, Ibadan 119, Oyo state, Nigeria
- Department of Biomedical Sciences, Faculty of Veterinary Sciences, University of Gadarif, Al-Gadarif 32211, Sudan
| | - Musa Ahmed
- Reproductive Health Sciences Program, Pan African University Life and Earth Sciences Institute, University of Ibadan, Ibadan 119, Oyo state, Nigeria
- Department of Obstetrics and Gynecology, College of Medicine, University of Ibadan, Ibadan 119, Oyo state, Nigeria
- Department of Veterinary Surgery, Faculty of Veterinary Medicine, AL-Salam University, Al-fula 120, West Kordofan, Sudan
| | - Adesina Oladokun
- Department of Obstetrics and Gynecology, College of Medicine, University of Ibadan, Ibadan 119, Oyo state, Nigeria
| |
Collapse
|
30
|
Van Der Kelen A, Okutman Ö, Javey E, Serdarogullari M, Janssens C, Ghosh MS, Dequeker BJH, Perold F, Kastner C, Kieffer E, Segers I, Gheldof A, Hes FJ, Sermon K, Verpoest W, Viville S. A systematic review and evidence assessment of monogenic gene-disease relationships in human female infertility and differences in sex development. Hum Reprod Update 2023; 29:218-232. [PMID: 36571510 DOI: 10.1093/humupd/dmac044] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Revised: 12/05/2022] [Indexed: 12/27/2022] Open
Abstract
BACKGROUND As in other domains of medicine, high-throughput sequencing methods have led to the identification of an ever-increasing number of gene variants in the fields of both male and female infertility. The increasing number of recently identified genes allows an accurate diagnosis for previously idiopathic cases of female infertility and more appropriate patient care. However, robust evidence of the gene-disease relationships (GDR) allowing the proper translation to clinical application is still missing in many cases. OBJECTIVE AND RATIONALE An evidence-based curation of currently identified genes involved in female infertility and differences in sex development (DSD) would significantly improve both diagnostic performance and genetic research. We therefore performed a systematic review to summarize current knowledge and assess the available GDR. SEARCH METHODS PRISMA guidelines were applied to curate all available information from PubMed and Web of Science on genetics of human female infertility and DSD leading to infertility, from 1 January 1988 to 1 November 2021. The reviewed pathologies include non-syndromic as well as syndromic female infertility, and endocrine and reproductive system disorders. The evidence that an identified phenotype is caused by pathogenic variants in a specific gene was assessed according to a standardized scoring system. A final score (no evidence, limited, moderate, strong, or definitive) was assigned to every GDR. OUTCOMES A total of 45 271 publications were identified and screened for inclusion of which 1078 were selected for gene and variant extraction. We have identified 395 genes and validated 466 GDRs covering all reported monogenic causes of female infertility and DSD. Furthermore, we present a genetic diagnostic flowchart including 105 genes with at least moderate evidence for female infertility and suggest recommendations for future research. The study did not take into account associated genetic risk factor(s) or oligogenic/polygenic causes of female infertility. WIDER IMPLICATIONS We have comprehensively reviewed the existing research on the genetics of female infertility and DSD, which will enable the development of diagnostic panels using validated genes. Whole genome analysis is shifting from predominantly research to clinical application, increasing its diagnostic potential. These new diagnostic possibilities will not only decrease the number of idiopathic cases but will also render genetic counselling more effective for infertile patients and their families.
Collapse
Affiliation(s)
- Annelore Van Der Kelen
- Clinical Sciences, Research Group Reproduction and Genetics, Centre for Medical Genetics, Vrije Universiteit Brussel (VUB), Universitair Ziekenhuis Brussel (UZ Brussel), Brussels, Belgium
| | - Özlem Okutman
- Laboratoire de Génétique Médicale LGM, Institut de Génétique Médicale d'Alsace IGMA, INSERM UMR 1112, Université de Strasbourg, Strasbourg, France.,Laboratoire de Diagnostic Génétique, Unité de Génétique de l'infertilité (UF3472), Hôpitaux Universitaires de Strasbourg, Strasbourg, France
| | - Elodie Javey
- Laboratoires de Diagnostic Génétique, Hôpitaux Universitaires de Strasbourg, Strasbourg, France
| | - Münevver Serdarogullari
- Department of Histology and Embryology, Faculty of Medicine, Cyprus International University, Northern Cyprus via Mersin 10, Turkey
| | - Charlotte Janssens
- Research Group Reproduction and Genetics, Vrije Universiteit Brussel (VUB), Brussels, Belgium
| | - Manjusha S Ghosh
- Research Group Reproduction and Genetics, Vrije Universiteit Brussel (VUB), Brussels, Belgium
| | - Bart J H Dequeker
- Clinical Sciences, Research Group Reproduction and Genetics, Centre for Medical Genetics, Vrije Universiteit Brussel (VUB), Universitair Ziekenhuis Brussel (UZ Brussel), Brussels, Belgium
| | - Florence Perold
- Research Group Reproduction and Genetics, Vrije Universiteit Brussel (VUB), Brussels, Belgium
| | - Claire Kastner
- Institut de Génétique Médicale d'Alsace IGMA, Hôpitaux Universitaires de Strasbourg, Strasbourg, France
| | - Emmanuelle Kieffer
- Service de Génétique Médicale, Laboratoires de Diagnostic Génétique, Unité de Diagnostic Préimplantatoire (UF9327), Hôpitaux Universitaires de Strasbourg, Strasbourg, France
| | - Ingrid Segers
- Clinical Sciences, Research Group Reproduction and Genetics, Brussels IVF Centre for Reproductive Medicine, Vrije Universiteit Brussel (VUB), Universitair Ziekenhuis Brussel (UZ Brussel), Brussels, Belgium.,Research Group Follicle Biology Laboratory (FOBI), Vrije Universiteit Brussel (VUB), Brussels, Belgium
| | - Alexander Gheldof
- Clinical Sciences, Research Group Reproduction and Genetics, Centre for Medical Genetics, Vrije Universiteit Brussel (VUB), Universitair Ziekenhuis Brussel (UZ Brussel), Brussels, Belgium
| | - Frederik J Hes
- Clinical Sciences, Research Group Reproduction and Genetics, Centre for Medical Genetics, Vrije Universiteit Brussel (VUB), Universitair Ziekenhuis Brussel (UZ Brussel), Brussels, Belgium
| | - Karen Sermon
- Research Group Reproduction and Genetics, Vrije Universiteit Brussel (VUB), Brussels, Belgium
| | - Willem Verpoest
- Clinical Sciences, Research Group Reproduction and Genetics, Brussels IVF Centre for Reproductive Medicine, Vrije Universiteit Brussel (VUB), Universitair Ziekenhuis Brussel (UZ Brussel), Brussels, Belgium
| | - Stéphane Viville
- Laboratoire de Génétique Médicale LGM, Institut de Génétique Médicale d'Alsace IGMA, INSERM UMR 1112, Université de Strasbourg, Strasbourg, France.,Laboratoire de Diagnostic Génétique, Unité de Génétique de l'infertilité (UF3472), Hôpitaux Universitaires de Strasbourg, Strasbourg, France
| |
Collapse
|
31
|
Solanki S, Kumar V, Kashyap P, Kumar R, De S, Datta TK. Beta-defensins as marker for male fertility: a comprehensive review†. Biol Reprod 2023; 108:52-71. [PMID: 36322147 DOI: 10.1093/biolre/ioac197] [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: 06/29/2022] [Revised: 10/15/2022] [Accepted: 10/21/2022] [Indexed: 12/05/2022] Open
Abstract
Bovine male fertility in animals has a direct impact on the productivity of dairy herds. The epididymal sperm maturations involve extensive sperm surface modifications to gain the fertilizing ability, especially by absorptions of the plethora of biomolecules, including glycoprotein beta-defensins (BDs), enzymes, organic ions, protein, and phospholipids. Defensins are broad-range nonspecific antimicrobial peptides that exhibit strong relations with innate and adaptive immunity, but their roles in male fertility are relatively recently identified. In the course of evolution, BD genes give rise to different clusters with specific functions, especially reproductive functions, by undergoing duplications and nonsynonymous mutations. BD polymorphisms have been reported with milk compositions, disease resistance, and antimicrobial activities. However, in recent decades, the link of BD polymorphisms with fertility has emerged as an appealing improvement of reproductive performance such as sperm motility, membrane integrity, cervical mucus penetration, evading of uterus immunosurveillance, oviduct cell attachment, and egg recognition. The reproductive-specific glycosylated BD class-A BDs (CA-BDs) have shown age- and sex-specific expressions in male reproductive organs, signifying their physiological pleiotropism, especially in the sperm maturation and sperm transport in the female reproductive tract. By considering adult male reproductive organ-specific BD expressions, importance in sperm functionalities, and bioinformatic analysis, we have selected two bovine BBD126 and BBD129 genes as novel potential biomarkers of bovine male fertility. Despite the importance of BDs, however, genomic characterization of most BD genes across most livestock and nonmodel organisms remains predictive/incomplete. The current review discusses our understanding of BD pleiotropic functions, polymorphism, and genomic structural attributes concerning the fertilizability of the male gamete in dairy animals.
Collapse
Affiliation(s)
- Subhash Solanki
- Animal Genomics Lab, National Dairy Research Institute, Karnal, India
| | - Vijay Kumar
- NMR lab-II, National Institute of immunology, New Delhi, India
| | - Poonam Kashyap
- Animal Genomics Lab, National Dairy Research Institute, Karnal, India
| | - Rakesh Kumar
- Animal Genomics Lab, National Dairy Research Institute, Karnal, India
| | - Sachinandan De
- Animal Genomics Lab, National Dairy Research Institute, Karnal, India
| | - Tirtha Kumar Datta
- Animal Genomics Lab, National Dairy Research Institute, Karnal, India.,ICAR- Central Institute for Research on Buffaloes, Hisar, India
| |
Collapse
|
32
|
Huang Y, Roig I. Genetic control of meiosis surveillance mechanisms in mammals. Front Cell Dev Biol 2023; 11:1127440. [PMID: 36910159 PMCID: PMC9996228 DOI: 10.3389/fcell.2023.1127440] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Accepted: 02/10/2023] [Indexed: 02/25/2023] Open
Abstract
Meiosis is a specialized cell division that generates haploid gametes and is critical for successful sexual reproduction. During the extended meiotic prophase I, homologous chromosomes progressively pair, synapse and desynapse. These chromosomal dynamics are tightly integrated with meiotic recombination (MR), during which programmed DNA double-strand breaks (DSBs) are formed and subsequently repaired. Consequently, parental chromosome arms reciprocally exchange, ultimately ensuring accurate homolog segregation and genetic diversity in the offspring. Surveillance mechanisms carefully monitor the MR and homologous chromosome synapsis during meiotic prophase I to avoid producing aberrant chromosomes and defective gametes. Errors in these critical processes would lead to aneuploidy and/or genetic instability. Studies of mutation in mouse models, coupled with advances in genomic technologies, lead us to more clearly understand how meiosis is controlled and how meiotic errors are linked to mammalian infertility. Here, we review the genetic regulations of these major meiotic events in mice and highlight our current understanding of their surveillance mechanisms. Furthermore, we summarize meiotic prophase genes, the mutations that activate the surveillance system leading to meiotic prophase arrest in mouse models, and their corresponding genetic variants identified in human infertile patients. Finally, we discuss their value for the diagnosis of causes of meiosis-based infertility in humans.
Collapse
Affiliation(s)
- Yan Huang
- Genome Integrity and Instability Group, Institut de Biotecnologia i Biomedicina, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Spain.,Histology Unit, Department of Cell Biology, Physiology, and Immunology, Cytology, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Spain
| | - Ignasi Roig
- Genome Integrity and Instability Group, Institut de Biotecnologia i Biomedicina, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Spain.,Histology Unit, Department of Cell Biology, Physiology, and Immunology, Cytology, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Spain
| |
Collapse
|
33
|
Batiha O, Burghel GJ, Alkofahi A, Alsharu E, Smith H, Alobaidi B, Al-Smadi M, Awamlah N, Hussein L, Abdelnour A, Sheth H, Veltman J. Screening by single-molecule molecular inversion probes targeted sequencing panel of candidate genes of infertility in azoospermic infertile Jordanian males. HUM FERTIL 2022; 25:939-946. [PMID: 34190021 PMCID: PMC7614817 DOI: 10.1080/14647273.2021.1946173] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Infertility is a common health problem that affects around 1 in 6 couples in the United States, where half of these cases are attributed to male factors. Genetics play an important role in infertility and it is estimated that up to 50% of cases are due to genetic factors. Despite this, many male infertility cases are still idiopathic. This study aimed to identify the presence of possibly pathogenic rare variants in a set of candidate genes related to azoospermia in 69 Jordanian men using a next-generation sequencing-based panel covering more than a hundred male infertility related genes. A total of 9 variants were found and validated. Among them, two variants included reported pathogenic variants in CFTR and one novel pathogenic variant in the USP9Y gene. We also report the detection of 6 other variants with uncertain significance in other genes. Interestingly, male cases with CFTR variants did not show the expected cystic fibrosis phenotypes except for infertility. This work helps to uncover the contribution of additional genetic factors to the aetiology of male infertility and highlights the importance to obtain more reliable information about the presence of genetic variation in the Jordanian population.
Collapse
Affiliation(s)
- Osamah Batiha
- Department of Biotechnology and Genetic Engineering, Jordan University of Science and Technology, Irbid, Jordan
| | - George J Burghel
- The Manchester Centre for Genomic Medicine, University of Manchester NHS Foundation Trust, Manchester, UK
| | - Ayesha Alkofahi
- Department of Biotechnology and Genetic Engineering, Jordan University of Science and Technology, Irbid, Jordan
| | - Emad Alsharu
- Reproductive Endocrinology and IVF Unit, King Hussein Medical Center, Amman, Jordan
| | - Hannah Smith
- Biosciences Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK
| | - Bilal Alobaidi
- Biosciences Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK
| | - Mohammad Al-Smadi
- Reproductive Endocrinology and IVF Unit, King Hussein Medical Center, Amman, Jordan
| | | | | | | | - Harsh Sheth
- Biosciences Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK.,FRIGE's Institute of Human Genetics, FRIGE House, Ahmedabad, India
| | - Joris Veltman
- Biosciences Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK
| |
Collapse
|
34
|
El-Dahtory F, Yahia S, Rasheed RA, Wahba Y. Prevalence and patterns of chromosomal abnormalities among Egyptian patients with infertility: a single institution’s 5-year experience. MIDDLE EAST FERTILITY SOCIETY JOURNAL 2022; 27:10. [DOI: 10.1186/s43043-022-00101-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Accepted: 04/11/2022] [Indexed: 11/10/2022] Open
Abstract
Abstract
Background
Chromosomal abnormalities represent an important cause of human infertility. Little is known about the prevalence of chromosomal abnormalities among Egyptian couples with infertility. We estimated the cytogenetic profiles and semen analysis patterns among infertile couples. We analyzed data from medical archives of 2150 patients with infertility in Mansoura University Children’s Hospital, Egypt from 2015 to 2019. The data included karyotypes and semen analysis reports.
Results
Chromosomal abnormalities were reported in 13.5% of infertile patients (290/2150); 150 out of 1290 (11.62%) males and 140 out of 860 (16.28%) females. Within the infertile males, the numerical chromosomal abnormalities were detected in 134/1290 (10.38%) males, and structural abnormalities were found in 16/1290 (1.24%) males. Within the infertile females, numerical sex chromosome abnormalities were detected in 75/860 (8.72%) females, structural sex chromosome abnormalities were found in 31/860 (3.6%) females, mosaicism of the sex chromosome was found in 22/860 (2.56%) females, and male pseudohermaphrodites were detected in 12/860 (1.39%) females.
Conclusions
Numerical chromosomal aberrations are the most frequent patterns among infertile couples. Attention should be paid to the traditional chromosomal analysis as an important diagnostic step in the infertility work-up.
Collapse
|
35
|
Thakur S, Sharma V, Kaur D, Purkait P. Angiotensin-Converting Enzyme (ACE) Insertion/Deletion (I/D) Polymorphism as a Conjoint Regulator of Coagulation, Fibrinolytic, and RAAS Pathway in Infertility and Associated Pregnancy Complications. J Renin Angiotensin Aldosterone Syst 2022; 2022:1695769. [PMID: 36532100 PMCID: PMC9726265 DOI: 10.1155/2022/1695769] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2022] [Revised: 08/31/2022] [Accepted: 09/14/2022] [Indexed: 10/29/2023] Open
Abstract
Despite the increase in assisted reproductive technologies, the high rates of infertility and pregnancy complications are a major concern to infertility specialists worldwide. Infertility may be attributed to pregnancy complications like thrombophilia, preeclampsia and fibrin-induced recurrent pregnancy loss (RPL). Renin-angiotensin-aldosterone system (RAAS) directly or indirectly causes preeclampsia and thrombophilia through the fibrinolytic pathway that ultimately leads to RPL or infertility. The underlying mechanisms of this interaction are still unclear. The present comprehensive review is intended to demonstrate the role and interaction of RAAS and fibrinolytic pathways in pregnancy complications. How this interaction can induce pregnancy complications, and ultimately infertility, is also discussed in the light of current evidence. This study also presents common markers that link RAAS and fibrinolytic processes in developing thrombophilia, preeclampsia and RPL. The common link in these pathways is ACE gene I/D polymorphism. Apart from ACE, PAI-1, VIIa, XIIa, AT1R, AT1AA, and TF are common molecules that can delineate the underlying causes of pregnancy complications and infertility.
Collapse
Affiliation(s)
- Sunil Thakur
- Origin LIFE Healthcare Solutions & Research Centre LLP, Chandigarh PIN-160036, India
| | - Vaishnavi Sharma
- Postgraduate Government College for Girls, Sector-42, Chandigarh, India
| | - Dipneet Kaur
- Origin LIFE Healthcare Solutions & Research Centre LLP, Chandigarh PIN-160036, India
| | - Pulakes Purkait
- Origin LIFE Healthcare Solutions & Research Centre LLP, Chandigarh PIN-160036, India
| |
Collapse
|
36
|
Wen L, Li G, Huang T, Geng W, Pei H, Yang J, Zhu M, Zhang P, Hou R, Tian G, Su W, Chen J, Zhang D, Zhu P, Zhang W, Zhang X, Zhang N, Zhao Y, Cao X, Peng G, Ren X, Jiang N, Tian C, Chen ZJ. Single-cell technologies: From research to application. Innovation (N Y) 2022; 3:100342. [PMID: 36353677 PMCID: PMC9637996 DOI: 10.1016/j.xinn.2022.100342] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Accepted: 10/13/2022] [Indexed: 11/09/2022] Open
Abstract
In recent years, more and more single-cell technologies have been developed. A vast amount of single-cell omics data has been generated by large projects, such as the Human Cell Atlas, the Mouse Cell Atlas, the Mouse RNA Atlas, the Mouse ATAC Atlas, and the Plant Cell Atlas. Based on these single-cell big data, thousands of bioinformatics algorithms for quality control, clustering, cell-type annotation, developmental inference, cell-cell transition, cell-cell interaction, and spatial analysis are developed. With powerful experimental single-cell technology and state-of-the-art big data analysis methods based on artificial intelligence, the molecular landscape at the single-cell level can be revealed. With spatial transcriptomics and single-cell multi-omics, even the spatial dynamic multi-level regulatory mechanisms can be deciphered. Such single-cell technologies have many successful applications in oncology, assisted reproduction, embryonic development, and plant breeding. We not only review the experimental and bioinformatics methods for single-cell research, but also discuss their applications in various fields and forecast the future directions for single-cell technologies. We believe that spatial transcriptomics and single-cell multi-omics will become the next booming business for mechanism research and commercial industry.
Collapse
Affiliation(s)
- Lu Wen
- Biomedical Pioneering Innovation Centre (BIOPIC), Peking University, Beijing 100871, China
| | - Guoqiang Li
- Biomedical Pioneering Innovation Centre (BIOPIC), Peking University, Beijing 100871, China
| | - Tao Huang
- Bio-Med Big Data Center, CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, Shanghai 200031, China
| | - Wei Geng
- School of Chemical Engineering and Technology, Sun Yat-Sen University, Zhuhai 519082, China
| | - Hao Pei
- Mozhuo Biotech (Zhejiang) Co., Ltd., Tongxiang, Jiaxing 314500, China
| | | | - Miao Zhu
- Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China
| | - Pengfei Zhang
- Department of Medical Oncology, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Rui Hou
- Geneis (Beijing) Co., Ltd., Beijing 100102, China
| | - Geng Tian
- Geneis (Beijing) Co., Ltd., Beijing 100102, China
| | - Wentao Su
- School of Food Science and Technology, Dalian Polytechnic University, Dalian 116034, China
| | - Jian Chen
- State Key Laboratory of Transducer Technology, Aerospace Information Research Institute, Chinese Academy of Sciences, Beijing 100190, China
| | - Dake Zhang
- Key Laboratory of Biomechanics and Mechanobiology, Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Engineering Medicine, Beihang University, Beijing 100083, China
| | - Pingan Zhu
- Department of Mechanical Engineering, City University of Hong Kong, Hong Kong 999077, China
| | - Wei Zhang
- Chongqing Institute of Green and Intelligent Technology, Chinese Academy of Sciences, Chongqing 400714, China
| | - Xiuxin Zhang
- Center of Peony, Institute of Vegetables and Flowers, Chinese Academy of Agricultural Sciences, Key Laboratory of Biology and Genetic Improvement of Flower Crops (North China), Key Laboratory of Biology and Genetic Improvement of Horticultural Crops, Ministry of Agriculture and Rural Affairs, Beijing 100081, China
| | - Ning Zhang
- Department of Hepatic Surgery, Fudan University Shanghai Cancer Center, Shanghai 200032, China
| | - Yunlong Zhao
- Advanced Technology Institute, University of Surrey, Guildford, Surrey, GU2 7XH, UK
- National Physical Laboratory, Teddington, Middlesex TW11 0LW, UK
| | - Xin Cao
- Institute of Clinical Science, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Guangdun Peng
- Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China
| | - Xianwen Ren
- Biomedical Pioneering Innovation Centre (BIOPIC), Peking University, Beijing 100871, China
| | - Nan Jiang
- West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu 610041, China
- Jinfeng Laboratory, Chongqing 401329, China
| | - Caihuan Tian
- Center of Peony, Institute of Vegetables and Flowers, Chinese Academy of Agricultural Sciences, Key Laboratory of Biology and Genetic Improvement of Flower Crops (North China), Key Laboratory of Biology and Genetic Improvement of Horticultural Crops, Ministry of Agriculture and Rural Affairs, Beijing 100081, China
| | - Zi-Jiang Chen
- Center for Reproductive Medicine, Shandong University, Jinan, Shandong, 250012, China
| |
Collapse
|
37
|
Fetal germ cell development in humans, a link with infertility. Semin Cell Dev Biol 2022; 131:58-65. [PMID: 35431137 DOI: 10.1016/j.semcdb.2022.03.035] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2021] [Revised: 03/26/2022] [Accepted: 03/29/2022] [Indexed: 12/14/2022]
Abstract
Gametes are cells that have the unique ability to give rise to new individuals as well as transmit (epi)genetic information across generations. Generation of functionally competent gametes, oocytes and sperm cells, depends to some extent on several fundamental processes that occur during fetal development. Direct studies on human fetal germ cells remain hindered by ethical considerations and inaccessibility to human fetal material. Therefore, the majority of our current knowledge of germ cell development still comes from an invaluable body of research performed using different mammalian species. During the last decade, our understanding of human fetal germ cells has increased due to the successful use of human pluripotent stem cells to model aspects of human early gametogenesis and advancements on single-cell omics. Together, this has contributed to determine the cell types and associated molecular signatures in the developing human gonads. In this review, we will put in perspective the knowledge obtained from several mammalian models (mouse, monkey, pig). Moreover, we will discuss the main events during human fetal (female) early gametogenesis and how the dysregulation of this highly complex and lengthy process can link to infertility later in life.
Collapse
|
38
|
Abu-Halima M, Becker LS, Ayesh BM, Meese E. MicroRNA-targeting in male infertility: Sperm microRNA-19a/b-3p and its spermatogenesis related transcripts content in men with oligoasthenozoospermia. Front Cell Dev Biol 2022; 10:973849. [PMID: 36211460 PMCID: PMC9533736 DOI: 10.3389/fcell.2022.973849] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Accepted: 09/08/2022] [Indexed: 11/21/2022] Open
Abstract
Objective: To elucidate and validate the potential regulatory function of miR-19a/b-3p and its spermatogenesis-related transcripts content in sperm samples collected from men with oligoasthenozoospermia. Methods: Men presenting at an infertility clinic were enrolled. MicroRNA (miRNA) and target genes evaluation were carried out using in silico prediction analysis, Reverse transcription-quantitative PCR (RT-qPCR) validation, and Western blot confirmation. Results: The expression levels of miRNA-19a/b-3p were significantly up-regulated and 51 target genes were significantly down-regulated in oligoasthenozoospermic men compared with age-matched normozoospermic men as determined by RT-qPCR. Correlation analysis highlighted that sperm count, motility, and morphology were negatively correlated with miRNA-19a/b-3p and positively correlated with the lower expression level of 51 significantly identified target genes. Furthermore, an inverse correlation between higher expression levels of miRNA-19a/b-3p and lower expression levels of 51 target genes was observed. Consistent with the results of the RT-qPCR, reduced expression levels of STK33 and DNAI1 protein levels were identified in an independent cohort of sperm samples collected from men with oligoasthenozoospermia. Conclusion: Findings suggest that the higher expression of miRNA-19a/b-3p or the lower expression of target genes are associated with oligoasthenozoospermia and male infertility, probably through influencing basic semen parameters. This study lay the groundwork for future studies focused on investigating therapies for male infertility.
Collapse
Affiliation(s)
| | | | - Basim M Ayesh
- Department of Laboratory Medical Sciences, Alaqsa University, Gaza, Palestine
| | - Eckart Meese
- Institute of Human Genetics, Saarland University, Homburg, Germany
| |
Collapse
|
39
|
Liu R, Yun Y, Shu W, Wang X, Luo M. Editorial: Reproductive genomics. Front Genet 2022; 13:1002458. [PMID: 36081991 PMCID: PMC9445836 DOI: 10.3389/fgene.2022.1002458] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Accepted: 07/28/2022] [Indexed: 11/13/2022] Open
Affiliation(s)
- Rong Liu
- Hubei Provincial Key Laboratory of Developmentally Originated Disease, Department of Human Histology and Embryology, Taikang Medical School (School of Basic Medical Sciences), Wuhan University, Wuhan, China
- *Correspondence: Rong Liu, ; Xi Wang, ; Mengcheng Luo,
| | - Yan Yun
- Department of Microbiology and Molecular Genetics, University of California, Davis, CA, United States
| | - Wenjie Shu
- Beijing Institute of Microbiology and Epidemiology, Beijing, China
| | - Xi Wang
- State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, China
- *Correspondence: Rong Liu, ; Xi Wang, ; Mengcheng Luo,
| | - Mengcheng Luo
- Hubei Provincial Key Laboratory of Developmentally Originated Disease, Department of Human Histology and Embryology, Taikang Medical School (School of Basic Medical Sciences), Wuhan University, Wuhan, China
- *Correspondence: Rong Liu, ; Xi Wang, ; Mengcheng Luo,
| |
Collapse
|
40
|
Sahota JS, Sharma B, Guleria K, Sambyal V. Candidate genes for infertility: an in-silico study based on cytogenetic analysis. BMC Med Genomics 2022; 15:170. [PMID: 35918717 PMCID: PMC9347124 DOI: 10.1186/s12920-022-01320-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Accepted: 07/22/2022] [Indexed: 11/26/2022] Open
Abstract
Background The cause of infertility remains unclear in a significant proportion of reproductive-age couples who fail to conceive naturally. Chromosomal aberrations have been identified as one of the main genetic causes of male and female infertility. Structural chromosomal aberrations may disrupt the functioning of various genes, some of which may be important for fertility. The present study aims to identify candidate genes and putative functional interaction networks involved in male and female infertility using cytogenetic data from cultured peripheral blood lymphocytes of infertile patients. Methods Karyotypic analyses was done in 201 infertile patients (100 males and 101 females) and 201 age and gender matched healthy controls (100 males and 101 females) after 72 h peripheral lymphocyte culturing and GTG banding, followed by bioinformatic analysis using Cytoscape v3.8.2 and Metascape. Results Several chromosomal regions with a significantly higher frequency of structural aberrations were identified in the infertile males (5q2, 10q2, and 17q2) and females (6q2, 16q2, and Xq2). Segregation of the patients based on type of infertility (primary v/s secondary infertility) led to the identification of chromosomal regions with a significantly higher frequency of structural aberrations exclusively within the infertile males (5q2, 17q2) and females (16q2) with primary infertility. Cytoscape identified two networks specific to these regions: a male specific network with 99 genes and a female specific network with 109 genes. The top enriched GO terms within the male and female infertility networks were “skeletal system morphogenesis” and “mRNA transport” respectively. PSME3, PSMD3, and CDC27 were the top 3 hub genes identified within the male infertility network. Similarly, UPF3B, IRF8, and PSMB1 were the top 3 hub genes identified with the female infertility network. Among the hub genes identified in the male- and female-specific networks, PSMB1, PSMD3, and PSME3 are functional components of the proteasome complex. These hub genes have a limited number of reports related to their respective roles in maintenance of fertility in mice model and humans and require validation in further studies. Conclusion The candidate genes predicted in the present study can serve as targets for future research on infertility. Supplementary Information The online version contains supplementary material available at 10.1186/s12920-022-01320-x.
Collapse
Affiliation(s)
- Jatinder Singh Sahota
- Department of Human Genetics, Cytogenetics Laboratory, Guru Nanak Dev University (GNDU), Amritsar, Punjab, 143005, India
| | - Bhavna Sharma
- Department of Human Genetics, Cytogenetics Laboratory, Guru Nanak Dev University (GNDU), Amritsar, Punjab, 143005, India
| | - Kamlesh Guleria
- Department of Human Genetics, Cytogenetics Laboratory, Guru Nanak Dev University (GNDU), Amritsar, Punjab, 143005, India
| | - Vasudha Sambyal
- Department of Human Genetics, Cytogenetics Laboratory, Guru Nanak Dev University (GNDU), Amritsar, Punjab, 143005, India.
| |
Collapse
|
41
|
Shetty S, Nair J, Johnson J, Shetty N, J AK, Thondehalmath N, Ganesh D, Bhat VR, M S, R A, Nayak R, Gunasheela D, Kadandale JS, Shetty S. Preimplantation Genetic Testing for Couples with Balanced Chromosomal Rearrangements. J Reprod Infertil 2022; 23:213-223. [PMID: 36415497 PMCID: PMC9666592 DOI: 10.18502/jri.v23i3.10013] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2021] [Accepted: 10/08/2021] [Indexed: 10/25/2023] Open
Abstract
BACKGROUND Chromosomal rearrangements play an important role in infertility. Carriers of chromosomal rearrangements have a lower chance of producing normal or balanced gametes due to abnormal segregation of chromosomes at meiosis, which leads to recurrent spontaneous abortions and infertility. Preimplantation genetic testing for structural chromosome rearrangements (PGT-SR) is offered to couples who have balanced chromosomal rearrangements in order to select embryos with a balanced karyotype prior to implantation, thereby increasing the chances of pregnancy. The purpose of the current study was to assess the outcomes of PGT-SR in patients carrying various balanced chromosomal rearrangements and to assess their clinical pregnancy outcome after in vitro fertilization (IVF). METHODS In this study, infertile couples with balanced chromosomal abnormalities undergoing PGT-SR were retrospectively analyzed at a single fertility center from January 2016 to December 2019. RESULTS PGT-SR was performed on 87 embryos from 22 couples in whom one partner carried a balanced translocation or an inversion. Fifty-seven (65.5%) of these embryos had unbalanced or sporadic aneuploidies, 30 (34.5%) embryos were normal or chromosomally balanced, which were then transferred in 18 couples. A higher rate of unbalanced translocations in comparison to sporadic aneuploidies was observed in couples with reciprocal translocation. The live birth rate per embryo transfer was found to be 66.6% (12/18). CONCLUSION PGT-SR is a useful tool in selecting normal or balanced embryos for transfer in IVF, which could lead to a pregnancy by reducing the chance of miscarriages due to chromosome aneuploidy in couples with balanced chromosomal rearrangements.
Collapse
Affiliation(s)
| | - Jiny Nair
- Tattvagene Pvt. Ltd., Bangalore, India
| | | | | | | | | | | | | | - Sajana M
- Gunasheela Surgical and Maternity Hospital, Bangalore, India
| | - Anjana R
- Gunasheela Surgical and Maternity Hospital, Bangalore, India
| | - Rajsekhar Nayak
- Tattvagene Pvt. Ltd., Bangalore, India
- Gunasheela Surgical and Maternity Hospital, Bangalore, India
| | - Devika Gunasheela
- Tattvagene Pvt. Ltd., Bangalore, India
- Gunasheela Surgical and Maternity Hospital, Bangalore, India
| | - Jayarama S Kadandale
- Tattvagene Pvt. Ltd., Bangalore, India
- Centre for Human Genetics Biotech Park, Bengaluru, India
| | - Swathi Shetty
- Tattvagene Pvt. Ltd., Bangalore, India
- Centre for Human Genetics Biotech Park, Bengaluru, India
| |
Collapse
|
42
|
FertilityOnline: A Straightforward Pipeline for Functional Gene Annotation and Disease Mutation Discovery. GENOMICS, PROTEOMICS & BIOINFORMATICS 2022; 20:455-465. [PMID: 34954426 PMCID: PMC9801063 DOI: 10.1016/j.gpb.2021.08.010] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/07/2019] [Revised: 06/02/2021] [Accepted: 09/27/2021] [Indexed: 01/26/2023]
Abstract
Exploring the genetic basis of human infertility is currently under intensive investigation. However, only a handful of genes have been validated in animal models as disease-causing genes in infertile men. Thus, to better understand the genetic basis of human spermatogenesis and bridge the knowledge gap between humans and other animal species, we construct the FertilityOnline, a database integrating the literature-curated functional genes during spermatogenesis into an existing spermatogenic database, SpermatogenesisOnline 1.0. Additional features, including the functional annotation and genetic variants of human genes, are also incorporated into FertilityOnline. By searching this database, users can browse the functional genes involved in spermatogenesis and instantly narrow down the number of candidates of genetic mutations underlying male infertility in a user-friendly web interface. Clinical application of this database was exampled by the identification of novel causative mutations in synaptonemal complex central element protein 1 (SYCE1) and stromal antigen 3 (STAG3) in azoospermic men. In conclusion, FertilityOnline is not only an integrated resource for spermatogenic genes but also a useful tool facilitating the exploration of the genetic basis of male infertility. FertilityOnline can be freely accessed at http://mcg.ustc.edu.cn/bsc/spermgenes2.0/index.html.
Collapse
|
43
|
Faridi R, Rea A, Fenollar-Ferrer C, O'Keefe RT, Gu S, Munir Z, Khan AA, Riazuddin S, Hoa M, Naz S, Newman WG, Friedman TB. New insights into Perrault syndrome, a clinically and genetically heterogeneous disorder. Hum Genet 2022; 141:805-819. [PMID: 34338890 PMCID: PMC11330641 DOI: 10.1007/s00439-021-02319-7] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Accepted: 07/14/2021] [Indexed: 01/07/2023]
Abstract
Hearing loss and impaired fertility are common human disorders each with multiple genetic causes. Sometimes deafness and impaired fertility, which are the hallmarks of Perrault syndrome, co-occur in a person. Perrault syndrome is inherited as an autosomal recessive disorder characterized by bilateral mild to severe childhood sensorineural hearing loss with variable age of onset in both sexes and ovarian dysfunction in females who have a 46, XX karyotype. Since the initial clinical description of Perrault syndrome 70 years ago, the phenotype of some subjects may additionally involve developmental delay, intellectual deficit and other neurological disabilities, which can vary in severity in part dependent upon the genetic variants and the gene involved. Here, we review the molecular genetics and clinical phenotype of Perrault syndrome and focus on supporting evidence for the eight genes (CLPP, ERAL1, GGPS1, HARS2, HSD17B4, LARS2, RMND1, TWNK) associated with Perrault syndrome. Variants of these eight genes only account for approximately half of the individuals with clinical features of Perrault syndrome where the molecular genetic base remains under investigation. Additional environmental etiologies and novel Perrault disease-associated genes remain to be identified to account for unresolved cases. We also report a new genetic variant of CLPP, computational structural insight about CLPP and single cell RNAseq data for eight reported Perrault syndrome genes suggesting a common cellular pathophysiology for this disorder. Some unanswered questions are raised to kindle future research about Perrault syndrome.
Collapse
Affiliation(s)
- Rabia Faridi
- Laboratory of Molecular Genetics, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Alessandro Rea
- Evolution and Genomic Sciences, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, M13 9PL, UK
- Manchester Centre for Genomic Medicine, Manchester University NHS Foundation Trust, Manchester, M13 9WL, UK
| | - Cristina Fenollar-Ferrer
- Laboratory of Molecular Genetics, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Raymond T O'Keefe
- Evolution and Genomic Sciences, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, M13 9PL, UK
- Manchester Centre for Genomic Medicine, Manchester University NHS Foundation Trust, Manchester, M13 9WL, UK
| | - Shoujun Gu
- Auditory Development and Restoration Program, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Zunaira Munir
- School of Biological Sciences, University of the Punjab, Quaid-i-Azam Campus, Lahore, 54590, Pakistan
- present address: Department of Neurosciences, University of Turin, 10124, Turin, Italy
| | - Asma Ali Khan
- Centre of Excellence in Molecular Biology, University of the Punjab, Lahore, 54000, Pakistan
| | - Sheikh Riazuddin
- Allama Iqbal Medical Research Center, Jinnah Burn and Reconstructive Surgery Center, University of Health Sciences, Lahore, 54550, Pakistan
| | - Michael Hoa
- Auditory Development and Restoration Program, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Sadaf Naz
- School of Biological Sciences, University of the Punjab, Quaid-i-Azam Campus, Lahore, 54590, Pakistan
| | - William G Newman
- Evolution and Genomic Sciences, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, M13 9PL, UK.
- Manchester Centre for Genomic Medicine, Manchester University NHS Foundation Trust, Manchester, M13 9WL, UK.
| | - Thomas B Friedman
- Laboratory of Molecular Genetics, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, MD, 20892, USA.
| |
Collapse
|
44
|
Behvarz M, Rahmani SA, Siasi Torbati E, Danaei Mehrabad S, Bikhof Torbati M. Association of CATSPER1, SPATA16 and TEX11 genes polymorphism with idiopathic azoospermia and oligospermia risk in Iranian population. BMC Med Genomics 2022; 15:47. [PMID: 35248021 PMCID: PMC8897944 DOI: 10.1186/s12920-022-01197-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Accepted: 02/25/2022] [Indexed: 11/10/2022] Open
Abstract
Abstract
Background
Male infertility is a heterogeneous disease which can occur due to spermatogenesis defects. The idiopathic azoospermia and oligospermia are the common cause of male infertility with unknown underlying molecular mechanisms. The aim of this study was to investigate association of idiopathic azoospermia and oligospermia with single-nucleotide polymorphisms of CATSPER1, SPATA16 and TEX11 genes in Iranian-Azeri men.
Methods
In this case–control study, we recruited 100 infertile men (case group) and 100 fertile men (control group) from Azeri population in north western provinces, Iran, population. The genomic DNA was extracted using a proteinase K method from peripheral blood leukocytes. The genotypes analysis was conducted using tetra-primer amplification refractory mutation system-polymerase chain reaction method. The obtained data were analyzed by statistical software.
Results
We found a significant difference in the frequencies of heterozygote AB and mutant homozygote BB genotypes in the CATSPER1 (rs2845570) gene polymorphism between patients and healthy controls (p < 0.05). Moreover, we observed a significant difference in the frequencies of heterozygote BA genotype in the SPATA16 (rs1515442) gene polymorphism between patients and healthy controls (p < 0.05). However, no significant difference was found in genotypes distribution of case and control groups in the TEX11 (rs143246552) gene polymorphism.
Conclusion
Our finding showed that the CATSPER1 (rs2845570) and SPATA16 (rs1515442) genes polymorphism may play an important role in idiopathic azoospermia and oligospermia in Iranian Azeri population. However, more extensive studies with larger sample sizes from different ethnic origins are essential for access more accurate results.
Collapse
|
45
|
Association of X Chromosome Aberrations with Male Infertility. ACTA MEDICA BULGARICA 2021. [DOI: 10.2478/amb-2021-0051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Abstract
Male infertility is caused by spermatogenetic failure, clinically noted as oligoor azoospermia. Approximately 20% of infertile patients carry a genetic defect. The most frequent genetic defect leading to azoospermia (or severe oligozoospermia) is Klinefelter syndrome (47, XXY), which is numerical chromosomal abnormality and Y- structural chromosome aberration. The human X chromosome is the most stable of all human chromosomes. The X chromosome is loaded with regions of acquired, rapidly evolving genes. The X chromosome may actually play an essential role in male infertility and sperm production. Here we will describe X chromosome aberrations, which are associated with male infertility.
Collapse
|
46
|
Wyrwoll MJ, van Walree ES, Hamer G, Rotte N, Motazacker MM, Meijers-Heijboer H, Alders M, Meißner A, Kaminsky E, Wöste M, Krallmann C, Kliesch S, Hunt TJ, Clark AT, Silber S, Stallmeyer B, Friedrich C, van Pelt AMM, Mathijssen IB, Tüttelmann F. Bi-allelic variants in DNA mismatch repair proteins MutS Homolog MSH4 and MSH5 cause infertility in both sexes. Hum Reprod 2021; 37:178-189. [PMID: 34755185 DOI: 10.1093/humrep/deab230] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Revised: 09/27/2021] [Indexed: 11/14/2022] Open
Abstract
STUDY QUESTION Do bi-allelic variants in the genes encoding the MSH4/MSH5 heterodimer cause male infertility? SUMMARY ANSWER We detected biallelic, (likely) pathogenic variants in MSH5 (4 men) and MSH4 (3 men) in six azoospermic men, demonstrating that genetic variants in these genes are a relevant cause of male infertility. WHAT IS KNOWN ALREADY MSH4 and MSH5 form a heterodimer, which is required for prophase of meiosis I. One variant in MSH5 and two variants in MSH4 have been described as causal for premature ovarian insufficiency (POI) in a total of five women, resulting in infertility. Recently, pathogenic variants in MSH4 have been reported in infertile men. So far, no pathogenic variants in MSH5 had been described in males. STUDY DESIGN, SIZE, DURATION We utilized exome data from 1305 men included in the Male Reproductive Genomics (MERGE) study, including 90 males with meiotic arrest (MeiA). Independently, exome sequencing was performed in a man with MeiA from a large consanguineous family. PARTICIPANTS/MATERIALS, SETTING, METHODS Assuming an autosomal-recessive mode of inheritance, we screened the exome data for rare, biallelic coding variants in MSH4 and MSH5. If possible, segregation analysis in the patients' families was performed. The functional consequences of identified loss-of-function (LoF) variants in MSH5 were studied using heterologous expression of the MSH5 protein in HEK293T cells. The point of arrest during meiosis was determined by γH2AX staining. MAIN RESULTS AND THE ROLE OF CHANCE We report for the first time (likely) pathogenic, homozygous variants in MSH5 causing infertility in 2 out of 90 men with MeiA and overall in 4 out of 902 azoospermic men. Additionally, we detected biallelic variants in MSH4 in two men with MeiA and in the sister of one proband with POI. γH2AX staining revealed an arrest in early prophase of meiosis I in individuals with pathogenic MSH4 or MSH5 variants. Heterologous in vitro expression of the detected LoF variants in MSH5 showed that the variant p.(Ala620GlnTer9) resulted in MSH5 protein truncation and the variant p.(Ser26GlnfsTer42) resulted in a complete loss of MSH5. LARGE SCALE DATA All variants have been submitted to ClinVar (SCV001468891-SCV001468896 and SCV001591030) and can also be accessed in the Male Fertility Gene Atlas (MFGA). LIMITATIONS, REASONS FOR CAUTION By selecting for variants in MSH4 and MSH5, we were able to determine the cause of infertility in six men and one woman, leaving most of the examined individuals without a causal diagnosis. WIDER IMPLICATIONS OF THE FINDINGS Our findings have diagnostic value by increasing the number of genes associated with non-obstructive azoospermia with high clinical validity. The analysis of such genes has prognostic consequences for assessing whether men with azoospermia would benefit from a testicular biopsy. We also provide further evidence that MeiA in men and POI in women share the same genetic causes. STUDY FUNDING/COMPETING INTEREST(S) This study was carried out within the frame of the German Research Foundation sponsored Clinical Research Unit 'Male Germ Cells: from Genes to Function' (DFG, CRU326), and supported by institutional funding of the Research Institute Amsterdam Reproduction and Development and funds from the LucaBella Foundation. The authors declare no conflict of interest.
Collapse
Affiliation(s)
- M J Wyrwoll
- Institute of Reproductive Genetics, University of Münster, Münster, Germany.,Department of Clinical and Surgical Andrology, Centre of Reproductive Medicine and Andrology, University Hospital Münster, Münster, Germany
| | - E S van Walree
- Department of Clinical Genetics, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands.,Department of Complex Trait Genetics, Center for Neurogenomics and Cognitive Research, Amsterdam Neuroscience, VU University, Amsterdam, The Netherlands
| | - G Hamer
- Reproductive Biology Laboratory, Center for Reproductive Medicine, Research Institute Amsterdam Reproduction and Development, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - N Rotte
- Institute of Reproductive Genetics, University of Münster, Münster, Germany
| | - M M Motazacker
- Laboratory of Genome Diagnostics, Department of Clinical Genetics, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - H Meijers-Heijboer
- Department of Clinical Genetics, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands.,Department of Clinical Genetics, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - M Alders
- Department of Clinical Genetics, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - A Meißner
- Department of Obstetrics and Gynecology, Center for Reproductive Medicine, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - E Kaminsky
- Praxis für Humangenetik, Hamburg, Germany
| | - M Wöste
- Institute of Medical Informatics, University of Münster, Münster, Germany
| | - C Krallmann
- Department of Clinical and Surgical Andrology, Centre of Reproductive Medicine and Andrology, University Hospital Münster, Münster, Germany
| | - S Kliesch
- Department of Clinical and Surgical Andrology, Centre of Reproductive Medicine and Andrology, University Hospital Münster, Münster, Germany
| | - T J Hunt
- Department of Molecular, Cell and Developmental Biology, Los Angeles, CA, USA
| | - A T Clark
- Department of Molecular, Cell and Developmental Biology, Los Angeles, CA, USA
| | - S Silber
- Infertility Center of St Louis, St Luke's Hospital, St Louis, MO, USA
| | - B Stallmeyer
- Institute of Reproductive Genetics, University of Münster, Münster, Germany
| | - C Friedrich
- Institute of Reproductive Genetics, University of Münster, Münster, Germany
| | - A M M van Pelt
- Reproductive Biology Laboratory, Center for Reproductive Medicine, Research Institute Amsterdam Reproduction and Development, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - I B Mathijssen
- Department of Clinical Genetics, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - F Tüttelmann
- Institute of Reproductive Genetics, University of Münster, Münster, Germany
| |
Collapse
|
47
|
Huang Z, Chen F, Xie M, Zhang H, Zhuang Y, Huang C, Li X, Liu H, Chen Z. The I510V mutation in KLHL10 in a patient with oligoasthenoteratozoospermia. J Reprod Dev 2021; 67:313-318. [PMID: 34433733 PMCID: PMC8568611 DOI: 10.1262/jrd.2021-063] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Oligoasthenoteratozoospermia is a human infertility syndrome caused by defects in spermatogenesis, spermiogenesis, and sperm maturation, and its etiology remains unclear. Kelch-like 10
(KLHL10) is a component of ubiquitin ligase E3 10 (KLHL10) and plays an important role in male fertility. Deletion or mutation of the Klhl10 gene in
Drosophila or mice results in defects in spermatogenesis or sperm maturation. However, the molecular mechanisms by which KLHL10 functions remain elusive. In this study, we
identified a missense mutation (c.1528A→G, p.I510V) in exon 5 of KLHL10, which is associated with oligoasthenoteratozoospermia in humans. To investigate the effects of this
mutation on KLHL10 function and spermatogenesis and/or spermiogenesis, we generated mutant mice duplicating the amino acid conversion using the clustered regularly interspaced palindromic
repeat/caspase 9 (CRISPR/Cas9) system and designated them Klhl10I510V mice. However, the Klhl10I510V mice did not exhibit any defects in testis development,
spermatogenesis, or sperm motility at ten-weeks-of-age, suggesting that this mutation does not disrupt the KLHL10 function, and may not be the cause of male infertility in the affected
individual with oligoasthenoteratozoospermia.
Collapse
Affiliation(s)
- Zicong Huang
- Department of Cell Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, P. R. China
| | - Feilong Chen
- Department of Pathology, Panyu Maternal and Child Care Service Centre of Guangzhou, Guangzhou 511499, P. R. China
| | - Minyu Xie
- Department of Cell Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, P. R. China
| | - Hanbin Zhang
- Department of Cell Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, P. R. China
| | - Yuge Zhuang
- Department of Cell Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, P. R. China
| | - Chuyu Huang
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, P. R. China
| | - Xuemei Li
- Reproductive center, Affiliated Shenzhen Maternity & Child Healthcare Hospital, Southern Medical University (Shenzhen Maternity & Child Healthcare Hospital), Shenzhen 518017, P. R. China
| | - Hong Liu
- Reproductive center, Affiliated Shenzhen Maternity & Child Healthcare Hospital, Southern Medical University (Shenzhen Maternity & Child Healthcare Hospital), Shenzhen 518017, P. R. China
| | - Zhenguo Chen
- Department of Cell Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, P. R. China
| |
Collapse
|
48
|
Wei X, Sha Y, Wei Z, Zhu X, He F, Zhang X, Liu W, Wang Y, Lu Z. Bi-allelic mutations in DNAH7 cause asthenozoospermia by impairing the integrality of axoneme structure. Acta Biochim Biophys Sin (Shanghai) 2021; 53:1300-1309. [PMID: 34476482 DOI: 10.1093/abbs/gmab113] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2021] [Indexed: 11/13/2022] Open
Abstract
Asthenozoospermia is the most common cause of male infertility. Dynein protein arms play a crucial role in the motility of both the cilia and flagella, and defects in these proteins generally impair the axoneme structure and cause primary ciliary dyskinesia. But relatively little is known about the influence of dynein protein arm defects on sperm flagella function. Here, we recruited 85 infertile patients with idiopathic asthenozoospermia and identified bi-allelic mutations in DNAH7 (NM_018897.3) from three patients using whole-exome sequencing. These variants are rare, highly pathogenic, and very conserved. The spermatozoa from the patients with DNAH7 bi-allelic mutations showed specific losses in the inner dynein arms. The expression of DNAH7 in the spermatozoa from the DNAH7-defective patients was significantly decreased, but these patients were able to have their children via intra-cytoplasmic sperm injection treatment. Our study is the first to demonstrate that bi-allelic mutations in DNAH7 may impair the integrality of axoneme structure, affect sperm motility, and cause asthenozoospermia in humans. These findings may extend the spectrum of etiological genes and provide new clues for the diagnosis and treatment of patients with asthenozoospermia.
Collapse
Affiliation(s)
- Xiaoli Wei
- School of Pharmaceutical Sciences, State Key Laboratory of Cellular Stress Biology, Xiamen University, Xiamen 361102, China
| | - Yanwei Sha
- Department of Andrology, United Diagnostic and Research Center for Clinical Genetics, Women and Children’s Hospital and School of Medicine, Xiamen University, Xiamen 361005, China
| | - Zijie Wei
- School of Pharmaceutical Sciences, State Key Laboratory of Cellular Stress Biology, Xiamen University, Xiamen 361102, China
| | - Xingshen Zhu
- School of Pharmaceutical Sciences, State Key Laboratory of Cellular Stress Biology, Xiamen University, Xiamen 361102, China
| | - Fengming He
- School of Pharmaceutical Sciences, State Key Laboratory of Cellular Stress Biology, Xiamen University, Xiamen 361102, China
| | - Xiaoya Zhang
- School of Pharmaceutical Sciences, State Key Laboratory of Cellular Stress Biology, Xiamen University, Xiamen 361102, China
| | - Wensheng Liu
- Obstetrics and Gynecology Center, Department of Obstetrics and Gynecology, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| | - Yifeng Wang
- Obstetrics and Gynecology Center, Department of Obstetrics and Gynecology, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| | - Zhongxian Lu
- School of Pharmaceutical Sciences, State Key Laboratory of Cellular Stress Biology, Xiamen University, Xiamen 361102, China
| |
Collapse
|
49
|
Eshghifar N, Dehghan BK, Do AA, Koukhaloo SZ, Habibi M, Pouresmaeili F. Infertility cell therapy and epigenetic insights. Hum Antibodies 2021; 29:17-26. [PMID: 33554898 DOI: 10.3233/hab-200438] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Recent advances in assisted reproductive technology (ART) have allowed couples with severe infertility to conceive, but the methods are not effective for all cases. Stem cells as undifferentiated cells which are found in different stages of embryonic, fetal and adult life are known to be capable of forming different cell types, tissues, and organs. Due to their unlimited resources and the incredible power of differentiation are considered as potential new therapeutic biological tools for treatment of infertility. For reproductive medicine, stem cells are stimulated in vitro to develop various specialized functional cells including male and female gametes. The epigenetic patterns can be modified in the genome under certain drugs exposure or lifestyle alterations. Therefore, epigenetics-related disorders may be treated if the nature of the modifications is completely admissible. It is proved that our understanding of epigenetic processes and its association with infertility would help us not only to understand the etiological factors but also to treat some type of male infertilities. Exploration of both genetic and epigenetic variations in the disease development could help in the identification of the interaction patterns between these two phenomena and possible improvement of therapeutic methods.
Collapse
Affiliation(s)
- Nahal Eshghifar
- Department of Cellular and Molecular Biology, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Behnam Kamali Dehghan
- Department of Medical Genetics, National Institute of Medical Engineering and Biotechnology (NIGEB), Tehran, Iran.,Medical Genetics, Jiroft University of Medical Sciences and Health Services, Jiroft, Kerman, Iran.,Department of Cellular and Molecular Biology, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Atieh Abedin Do
- Groupe de Recherche en Écologie Buccale, Faculté de Médecine Dentaire, Université Laval, Quebec, Canada
| | | | - Mohsen Habibi
- Department of Medical Genetics, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Farkhondeh Pouresmaeili
- Department of Medical Genetics, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
50
|
Faiza H, Khan M, Rafiq M, Khan AA, Rind NA, Naqvi SHA. Two novel mutations in exon 2 of bone morphogenetic protein (BMP) 15 gene in Pakistani infertile females. Saudi J Biol Sci 2021; 28:5364-5370. [PMID: 34466116 PMCID: PMC8381042 DOI: 10.1016/j.sjbs.2021.05.061] [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: 03/29/2021] [Revised: 05/23/2021] [Accepted: 05/24/2021] [Indexed: 12/01/2022] Open
Abstract
Objective To determine the proportion of fertility in Pakistani infertile females and discover if there are considerable connection among BMP15 gene polymorphism, follicle maturation and hormonal regulation in Pakistani infertile females. Methods All selected participants were initially examined through follicle-stimulating hormones (FSH), luteinizing hormone (LH), thyroid-stimulating hormone (TSH), Prolactin, and Trans-vaginal scan (TVS). BMP15 gene polymorphism among infertile and fertile females was done by extracted Genomic DNA from whole blood. Sanger sequencing was performed for the identification of mutation in exons-intron boundaries of the BMP15 gene. Bioinformatics tools were used to assess the protein structure. Results The total five mutations including two novel missense variants of BMP15 in exon 2, whereas three previously reported i.e. two cosmic mutations (c.615delC), (c.584InsG) and one frame shift mutations (c.635delA) were also observed. The first novel mutation was found at (c.1038InsGG) (p.346Gln < Gly) in which the insertion of GG at DNA position 1038 of exon 2 resulting in a substitution of glutamine into glycine at 346th amino acid of BMP15 protein. The second novel variant (c.1049delT) (p. Ser334Pro) was also observed in exon 2 of the BMP15 gene, which substituted serine into proline at 334th amino acid of the BMP15 protein. Conclusion It is concluded that there are various missense mutations present in exon 2 of the BMP15 gene of Pakistani infertile females, consequently expected function of protein changes due to change in codons of amino acids. Provean and SIFT suggest the two novel variants as potentially deleterious. Although three other variants were also found in Pakistani infertile females which were previously reported. These mutations may result in early blockage of folliculogenesis and ovaries become streaky. Further research is required to resolve the actual allusion of these variations in the BMP15 gene.
Collapse
Affiliation(s)
- Hafiza Faiza
- Institute of Biotechnology and Genetic Engineering, University of Sindh, Jamshoro, Pakistan
| | - Majida Khan
- Department of Gynecology, Liaquat University of Medical Science, Jamshoro, Pakistan
| | - Muhammad Rafiq
- Institute of Biotechnology and Genetic Engineering, University of Sindh, Jamshoro, Pakistan
| | - Anoshiya Ali Khan
- Institute of Biotechnology and Genetic Engineering, University of Sindh, Jamshoro, Pakistan
| | - Nadir Ali Rind
- Institute of Biotechnology and Genetic Engineering, University of Sindh, Jamshoro, Pakistan.,Department of Genetics & Molecular Biology, SBBU, Shaheed Benazirabad, Pakistan
| | - Syed Habib Ahmed Naqvi
- Institute of Biotechnology and Genetic Engineering, University of Sindh, Jamshoro, Pakistan
| |
Collapse
|