1
|
Qiu W, Yin S, Abulaiti K, Li X, Lu Y, Zhang Q, Zhan M, Zhang J. Preconception exposure to bisphenol A and its alternatives: Effects on female fecundity mediated by oxidative stress and ovarian reserve. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 957:177558. [PMID: 39547376 DOI: 10.1016/j.scitotenv.2024.177558] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Revised: 11/03/2024] [Accepted: 11/12/2024] [Indexed: 11/17/2024]
Abstract
BACKGROUND Various 'Bisphenol A (BPA)-free' alternatives have emerged in numerous personal products in recent years. However, it remains unclear whether BPA analogs affect female fecundity and possible biological mechanisms. OBJECTIVES We aimed to evaluate the relationships of bisphenol analogs with female fecundability and infertility and whether oxidative stress, inflammation, and ovarian reserve may play a mediation role in such associations. METHODS This prospective preconception cohort study included 957 couples who attempted pregnancy. BPA and six alternatives were measured in women's urine samples. Bisphenol analogs-outcome associations were estimated using discrete-time Cox hazards and logistic regression models. A quantile g-computation (QGC) methodology was further applied to assess the joint effects of co-exposure to bisphenol analogs on fecundity. We also quantified three biomarkers, including malondialdehyde (MDA), C-reactive protein and Anti-Müllerian hormone (AMH), to explore possible biological pathways. RESULTS Using an integrated analytical approach consisting of both single-pollutant and mixture models, we found that BPA and bisphenol AP (BPAP) were significantly associated with decreased fecundability (adjusted fecundability ratio (aFR) = 0.87, 95%CI: 0.81, 0.94 for BPA; aFR = 0.64, 95%CI: 0.48, 0.84 for BPAP) and increased risk of infertility (adjusted odd ratio (aOR) = 1.23, 95%CI: 1.06, 1.44 for BPA; aOR = 2.27, 95%CI: 1.29, 3.99 for BPAP) after controlling for other bisphenol analogs. The link between BPA and prolonged time to pregnancy was more prominent in overweight or obese women and those who had regular menstrual cycles. Bisphenol AF was associated with impaired fecundity in women aged 35 years or older. The mixed effects of bisphenol analogs on fecundity were statistically non-significant. Mediation analysis revealed a significant indirect effect of urinary MDA and serum AMH in bisphenol analogs-induced impaired fecundity, with all average causal mediation effects (ACME) showing statistical significance (PACME < 0.05). CONCLUSIONS Our prospective preconception cohort study suggests that BPA and BPAP may be associated with impaired female fecundity. Increased oxidative stress and decreased ovarian reserve may be the underlying pathways.
Collapse
Affiliation(s)
- Wei Qiu
- Ministry of Education-Shanghai Key Laboratory of Children's Environmental Health, School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Shengju Yin
- Ministry of Education-Shanghai Key Laboratory of Children's Environmental Health, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
| | - Kadila Abulaiti
- Ministry of Education-Shanghai Key Laboratory of Children's Environmental Health, School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Xinyu Li
- Center for Reproductive Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200135, China
| | - Yao Lu
- Center for Reproductive Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200135, China
| | - Qianlong Zhang
- Ministry of Education-Shanghai Key Laboratory of Children's Environmental Health, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
| | - Ming Zhan
- Pudong New Area Center for Disease Control and Prevention, Shanghai 200136, China.
| | - Jun Zhang
- Center for Reproductive Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200135, China.
| |
Collapse
|
2
|
Zeng Y, Liu W, Luo Y, Luo B, Zhu L, Yang Z, Feng K, Li D, Chen SA, Li X. The impact of Duostim protocol on pregnancy outcomes in infertile patients: A meta-analysis comparing single and double conventional stimulation cycles. J Assist Reprod Genet 2024:10.1007/s10815-024-03304-5. [PMID: 39601990 DOI: 10.1007/s10815-024-03304-5] [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: 07/30/2024] [Accepted: 10/21/2024] [Indexed: 11/29/2024] Open
Abstract
BACKGROUND The DuoStim protocol has been proposed as an alternative to conventional single and double stimulation cycles in the treatment of infertility. However, its efficacy in improving pregnancy outcomes remains uncertain. OBJECTIVE To systematically evaluate the impact of the DuoStim protocol on pregnancy outcomes in infertile patients by comparing it with single and double conventional stimulation cycles. METHODS An online systematic search was conducted using PubMed, Cochrane Library, and EMBASE databases, covering the period from their inception to March 2024. Randomized controlled trials (RCTs) comparing the DuoStim protocol with single and double conventional stimulation cycles in infertile patients were identified. Data were extracted by two independent investigators who screened the literature and assessed the quality of the studies. Meta-analysis was performed using RevMan 5.4 software. RESULTS A total of six RCTs involving 414 infertile patients were included. The DuoStim protocol significantly increased the total number of oocytes compared to single and double conventional stimulation (MD = - 1.47; 95% CI, - 2.12 to - 0.82; P < 0.00001). There were no statistically significant differences in the number of MII oocytes, total embryos, pregnancy rate, and live birth rate. Subgroup Analysis: compared to single stimulation, the DuoStim protocol significantly increased the number of MII oocytes (MD = 1.71; 95% CI, 0.77 to 2.66; P = 0.0004) and total embryos (MD = 1.34; 95% CI, 0.61 to 2.08; P = 0.0003). There were no significant differences in pregnancy rate and live birth rate. Secondary outcomes showed the effect of the DuoStim protocol in patients undergoing preimplantation genetic testing for aneuploidies (PGT-A). The time to obtain euploid blastocysts was significantly reduced in the DuoStim group compared to the control group (23.3 ± 2.8 days vs. 44.1 ± 2.0 days; P < 0.001). CONCLUSIONS The DuoStim protocol shows a significant advantage in increasing the total number of oocytes, MII oocytes, and embryos compared to single stimulation. However, it does not significantly improve pregnancy and live birth rates. The protocol also shortens the time to obtain euploid blastocysts in patients undergoing PGT-A, indicating potential benefits for specific patient groups. Further research is needed to confirm these findings and evaluate long-term outcomes. Thus, the quality of evidence should be considered moderate, warranting cautious interpretation of the results.
Collapse
Affiliation(s)
- Youman Zeng
- Finance Section, Maternal and Child Health Care Hospital of Yulin, No. 290, Qingning Road, Yulin Guangxi, 537000, China
| | - Weiwu Liu
- Reproductive Medicine Center, Maternal and Child Health Care Hospital of Yulin, No. 290, Qingning Road, Yulin Guangxi, 537000, China
| | - Yudi Luo
- Reproductive Medicine Center, Maternal and Child Health Care Hospital of Yulin, No. 290, Qingning Road, Yulin Guangxi, 537000, China
| | - Bowen Luo
- Reproductive Medicine Center, Maternal and Child Health Care Hospital of Yulin, No. 290, Qingning Road, Yulin Guangxi, 537000, China
| | - Lingling Zhu
- Reproductive Medicine Center, Maternal and Child Health Care Hospital of Yulin, No. 290, Qingning Road, Yulin Guangxi, 537000, China
| | - Zengyu Yang
- Reproductive Medicine Center, Maternal and Child Health Care Hospital of Yulin, No. 290, Qingning Road, Yulin Guangxi, 537000, China
| | - Keng Feng
- Reproductive Medicine Center, Maternal and Child Health Care Hospital of Yulin, No. 290, Qingning Road, Yulin Guangxi, 537000, China
| | - Derong Li
- Reproductive Medicine Center, Maternal and Child Health Care Hospital of Yulin, No. 290, Qingning Road, Yulin Guangxi, 537000, China
| | - Sheng-Ao Chen
- College of Animal Sciences, Xinjiang Uygur Autonomous Region 843300, Tarim University, Alar, China
| | - Xiang Li
- Reproductive Medicine Center, Maternal and Child Health Care Hospital of Yulin, No. 290, Qingning Road, Yulin Guangxi, 537000, China.
| |
Collapse
|
3
|
Guzmán-Jiménez A, González-Muñoz S, Cerván-Martín M, Garrido N, Castilla JA, Gonzalvo MC, Clavero A, Molina M, Luján S, Santos-Ribeiro S, Vilches MÁ, Espuch A, Maldonado V, Galiano-Gutiérrez N, Santamaría-López E, González-Ravina C, Quintana-Ferraz F, Gómez S, Amorós D, Martínez-Granados L, Ortega-González Y, Burgos M, Pereira-Caetano I, Bulbul O, Castellano S, Romano M, Albani E, Bassas L, Seixas S, Gonçalves J, Lopes AM, Larriba S, Palomino-Morales RJ, Carmona FD, Bossini-Castillo L. A comprehensive study of common and rare genetic variants in spermatogenesis-related loci identifies new risk factors for idiopathic severe spermatogenic failure. Hum Reprod Open 2024; 2024:hoae069. [PMID: 39678461 PMCID: PMC11645127 DOI: 10.1093/hropen/hoae069] [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: 07/26/2024] [Revised: 10/11/2024] [Indexed: 12/17/2024] Open
Abstract
STUDY QUESTION Can genome-wide genotyping data be analysed using a hypothesis-driven approach to enhance the understanding of the genetic basis of severe spermatogenic failure (SPGF) in male infertility? SUMMARY ANSWER Our findings revealed a significant association between SPGF and the SHOC1 gene and identified three novel genes (PCSK4, AP3B1, and DLK1) along with 32 potentially pathogenic rare variants in 30 genes that contribute to this condition. WHAT IS KNOWN ALREADY SPGF is a major cause of male infertility, often with an unknown aetiology. SPGF can be due to either multifactorial causes, including both common genetic variants in multiple genes and environmental factors, or highly damaging rare variants. Next-generation sequencing methods are useful for identifying rare mutations that explain monogenic forms of SPGF. Genome-wide association studies (GWASs) have become essential approaches for deciphering the intricate genetic landscape of complex diseases, offering a cost-effective and rapid means to genotype millions of genetic variants. Novel methods have demonstrated that GWAS datasets can be used to infer rare coding variants that are causal for male infertility phenotypes. However, this approach has not been previously applied to characterize the genetic component of a whole case-control cohort. STUDY DESIGN SIZE DURATION We employed a hypothesis-driven approach focusing on all genetic variation identified, using a GWAS platform and subsequent genotype imputation, encompassing over 20 million polymorphisms and a total of 1571 SPGF patients and 2431 controls. Both common (minor allele frequency, MAF > 0.01) and rare (MAF < 0.01) variants were investigated within a total of 1797 loci with a reported role in spermatogenesis. This gene panel was meticulously assembled through comprehensive searches in the literature and various databases focused on male infertility genetics. PARTICIPANTS/MATERIALS SETTING METHODS This study involved a European cohort using previously and newly generated data. Our analysis consisted of three independent methods: (i) variant-wise association analyses using logistic regression models, (ii) gene-wise association analyses using combined multivariate and collapsing burden tests, and (iii) identification and characterisation of highly damaging rare coding variants showing homozygosity only in SPGF patients. MAIN RESULTS AND THE ROLE OF CHANCE The variant-wise analyses revealed an association between SPGF and SHOC1-rs12347237 (P = 4.15E-06, odds ratio = 2.66), which was likely explained by an altered binding affinity of key transcription factors in regulatory regions and the disruptive effect of coding variants within the gene. Three additional genes (PCSK4, AP3B1, and DLK1) were identified as novel relevant players in human male infertility using the gene-wise burden test approach (P < 5.56E-04). Furthermore, we linked a total of 32 potentially pathogenic and recessive coding variants of the selected genes to 35 different cases. LARGE SCALE DATA Publicly available via GWAS catalog (accession number: GCST90239721). LIMITATIONS REASONS FOR CAUTION The analysis of low-frequency variants presents challenges in achieving sufficient statistical power to detect genetic associations. Consequently, independent studies with larger sample sizes are essential to replicate our results. Additionally, the specific roles of the identified variants in the pathogenic mechanisms of SPGF should be assessed through functional experiments. WIDER IMPLICATIONS OF THE FINDINGS Our findings highlight the benefit of using GWAS genotyping to screen for both common and rare variants potentially implicated in idiopathic cases of SPGF, whether due to complex or monogenic causes. The discovery of novel genetic risk factors for SPGF and the elucidation of the underlying genetic causes provide new perspectives for personalized medicine and reproductive counselling. STUDY FUNDING/COMPETING INTERESTS This work was supported by the Spanish Ministry of Science and Innovation through the Spanish National Plan for Scientific and Technical Research and Innovation (PID2020-120157RB-I00) and the Andalusian Government through the research projects of 'Plan Andaluz de Investigación, Desarrollo e Innovación (PAIDI 2020)' (ref. PY20_00212) and 'Proyectos de Investigación aplicada FEDER-UGR 2023' (ref. C-CTS-273-UGR23). S.G.-M. was funded by the previously mentioned projects (ref. PY20_00212 and PID2020-120157RB-I00). A.G.-J. was funded by MCIN/AEI/10.13039/501100011033 and FSE 'El FSE invierte en tu futuro' (grant ref. FPU20/02926). IPATIMUP integrates the i3S Research Unit, which is partially supported by the Portuguese Foundation for Science and Technology (FCT), financed by the European Social Funds (COMPETE-FEDER) and National Funds (projects PEstC/SAU/LA0003/2013 and POCI-01-0145-FEDER-007274). S.S. is supported by FCT funds (10.54499/DL57/2016/CP1363/CT0019), ToxOmics-Centre for Toxicogenomics and Human Health, Genetics, Oncology and Human Toxicology, and is also partially supported by the Portuguese Foundation for Science and Technology (UIDP/00009/2020 and UIDB/00009/2020). S. Larriba received support from Instituto de Salud Carlos III (grant: DTS18/00101), co-funded by FEDER funds/European Regional Development Fund (ERDF)-a way to build Europe) and from 'Generalitat de Catalunya' (grant 2021SGR052). S. Larriba is also sponsored by the 'Researchers Consolidation Program' from the SNS-Dpt. Salut Generalitat de Catalunya (Exp. CES09/020). All authors declare no conflict of interest related to this study.
Collapse
Affiliation(s)
- Andrea Guzmán-Jiménez
- Departamento de Genética e Instituto de Biotecnología, Centro de Investigación Biomédica (CIBM), Universidad de Granada, Granada, Spain
- Instituto de Investigación Biosanitaria ibs. GRANADA, Granada, Spain
| | - Sara González-Muñoz
- Departamento de Genética e Instituto de Biotecnología, Centro de Investigación Biomédica (CIBM), Universidad de Granada, Granada, Spain
- Instituto de Investigación Biosanitaria ibs. GRANADA, Granada, Spain
| | - Miriam Cerván-Martín
- Institute of Parasitology and Biomedicine López-Neyra (IPBLN), CSIC, Granada, Spain
| | - Nicolás Garrido
- IVIRMA Global Research Alliance, IVI Foundation, Instituto de Investigación Sanitaria La Fe (IIS La Fe), Valencia, Spain
| | - José A Castilla
- Instituto de Investigación Biosanitaria ibs. GRANADA, Granada, Spain
- Departamento de Anatomía y Embriología Humana, Facultad de Medicina, Universidad de Granada, Granada, Spain
| | - M Carmen Gonzalvo
- Instituto de Investigación Biosanitaria ibs. GRANADA, Granada, Spain
- Unidad de Reproducción, UGC Obstetricia y Ginecología, HU Virgen de las Nieves, Granada, Spain
| | - Ana Clavero
- Instituto de Investigación Biosanitaria ibs. GRANADA, Granada, Spain
- Unidad de Reproducción, UGC Obstetricia y Ginecología, HU Virgen de las Nieves, Granada, Spain
| | - Marta Molina
- Instituto de Investigación Biosanitaria ibs. GRANADA, Granada, Spain
- Unidad de Reproducción, UGC Obstetricia y Ginecología, HU Virgen de las Nieves, Granada, Spain
| | - Saturnino Luján
- Servicio de Urología, Hospital Universitari i Politecnic La Fe e Instituto de Investigación Sanitaria La Fe (IIS La Fe), Valencia, Spain
| | - Samuel Santos-Ribeiro
- IVI-RMA Lisbon, Lisbon, Portugal
- Department of Obstetrics and Gynecology, Faculty of Medicine, University of Lisbon, Lisbon, Portugal
| | - Miguel Ángel Vilches
- Ovoclinic & Ovobank, Clínicas de Reproducción Asistida y Banco de óvulos, Marbella, Málaga, Spain
| | - Andrea Espuch
- Hospital Universitario Torrecárdenas, Unidad de Reproducción Humana Asistida, Almería, Spain
| | - Vicente Maldonado
- UGC de Obstetricia y Ginecología, Complejo Hospitalario de Jaén, Jaén, Spain
| | | | | | - Cristina González-Ravina
- IVIRMA Global Research Alliance, IVI Foundation, Instituto de Investigación Sanitaria La Fe (IIS La Fe), Valencia, Spain
| | - Fernando Quintana-Ferraz
- IVIRMA Global Research Alliance, IVI Foundation, Instituto de Investigación Sanitaria La Fe (IIS La Fe), Valencia, Spain
| | - Susana Gómez
- IVIRMA Global Research Alliance, IVI Foundation, Instituto de Investigación Sanitaria La Fe (IIS La Fe), Valencia, Spain
| | - David Amorós
- IVIRMA Global Research Alliance, IVI Foundation, Instituto de Investigación Sanitaria La Fe (IIS La Fe), Valencia, Spain
| | | | | | - Miguel Burgos
- Departamento de Genética e Instituto de Biotecnología, Centro de Investigación Biomédica (CIBM), Universidad de Granada, Granada, Spain
| | - Iris Pereira-Caetano
- Departamento de Genética Humana, Instituto Nacional de Saúde Dr Ricardo Jorge, Lisbon, Portugal
| | - Ozgur Bulbul
- Division of Gynecology and Reproductive Medicine, Department of Gynecology, Fertility Center, Humanitas Research Hospital, IRCCS, Milan, Italy
| | - Stefano Castellano
- Division of Gynecology and Reproductive Medicine, Department of Gynecology, Fertility Center, Humanitas Research Hospital, IRCCS, Milan, Italy
| | - Massimo Romano
- Division of Gynecology and Reproductive Medicine, Department of Gynecology, Fertility Center, Humanitas Research Hospital, IRCCS, Milan, Italy
- Department of Biomedical Sciences, Humanitas University, Milan, Italy
| | - Elena Albani
- Division of Gynecology and Reproductive Medicine, Department of Gynecology, Fertility Center, Humanitas Research Hospital, IRCCS, Milan, Italy
| | - Lluís Bassas
- Laboratory of Seminology and Embryology, Andrology Service-Fundació Puigvert, Barcelona, Spain
| | - Susana Seixas
- i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- Institute of Molecular Pathology and Immunology of the University of Porto (IPATIMUP), Porto, Portugal
| | - João Gonçalves
- Departamento de Genética Humana, Instituto Nacional de Saúde Dr Ricardo Jorge, Lisbon, Portugal
- ToxOmics—Centro de Toxicogenómica e Saúde Humana, Nova Medical School, Lisbon, Portugal
| | - Alexandra M Lopes
- Institute of Molecular Pathology and Immunology of the University of Porto (IPATIMUP), Porto, Portugal
- CGPP-IBMC—Centro de Genética Preditiva e Preventiva, Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, Portugal
| | - Sara Larriba
- Human Molecular Genetics Group, Bellvitge Biomedical Research Institute (IDIBELL), L’Hospitalet de Llobregat, Barcelona, Spain
| | - Rogelio J Palomino-Morales
- Instituto de Investigación Biosanitaria ibs. GRANADA, Granada, Spain
- Departamento de Bioquímica y Biología Molecular I, Universidad de Granada, Granada, Spain
| | - F David Carmona
- Departamento de Genética e Instituto de Biotecnología, Centro de Investigación Biomédica (CIBM), Universidad de Granada, Granada, Spain
- Instituto de Investigación Biosanitaria ibs. GRANADA, Granada, Spain
| | - Lara Bossini-Castillo
- Departamento de Genética e Instituto de Biotecnología, Centro de Investigación Biomédica (CIBM), Universidad de Granada, Granada, Spain
- Instituto de Investigación Biosanitaria ibs. GRANADA, Granada, Spain
| |
Collapse
|
4
|
Lin Y, Wei Z, Zhang L, Yao Y, Huang Y, Yao G, Wang W, Hu S, Ding Y, Lu Y, Bian X, Dong X, Guan H, Huang Y, Sun Y. Homozygous missense variations of APC12 cause meiotic metaphase I arrest in oocytes and female infertility. Am J Obstet Gynecol 2024:S0002-9378(24)01140-2. [PMID: 39542389 DOI: 10.1016/j.ajog.2024.11.013] [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: 06/17/2024] [Revised: 10/25/2024] [Accepted: 11/05/2024] [Indexed: 11/17/2024]
Abstract
BACKGROUND Oocyte maturation arrest is a leading cause of female infertility. However, the genetic variables remain largely unknown. In oocytes, the activation of anaphase-promoting complex/cyclosome (APC/C) is a critical step in the transition from meiosis I to meiosis II. However, the causal relationship between variants in APC/C components and female infertility has not been completely investigated. OBJECTIVE This study aims to find a novel gene and its pathogenic mutation as a cause for metaphase I arrest in oocytes, thus introducing a new APC/C component for screening causes of female infertility. STUDY DESIGN Whole-exome sequencing was performed on 30 infertile women with recurrent oocyte maturation arrest without known gene variants. A homozygous missense mutation in the APC12 gene (p.R8H) was identified as a candidate for oocyte metaphase I arrest in a consanguineous family member. The experiment was conducted in vitro with HEK293T cells and mouse oocytes. Methods such as oocyte microinjection, western blotting, co-immunoprecipitation, and immunofluorescence were used. A knockdown mouse model was generated to verify the function of Apc12 in causing oocyte metaphase I arrest. About 100 wild-type C57BL/6J mice and 50 gene-edited mice were used in this study. RESULTS APC12 p.R8H was identified in an infertile woman with oocyte metaphase I arrest. Microinjection of Apc12 mutant cRNA in mouse oocytes caused a considerably higher rate of metaphase I stage arrest (65.21±5.64% vs 30.86±1.74%, P<.01) with decreased APC12 protein expression and Securin accumulation compared to the control group, while oocytes injected with Apc12 cRNA showed comparable metaphase I arrest rate (31.51±3.05%). This fit the phenotype we identified in our case and suggested that Apc12 p.R8H was a loss-of-function mutation leading to oocyte metaphase I arrest. The in vitro experiments in HEK293T cells suggested that the APC12 p.R8H mutation disrupted the interaction between APC12 and APC6, as well as impaired APC/C activity by disrupting Securin ubiquitination. Knocking down APC12 with siRNA in mouse oocytes led to metaphase I arrest (41.65±6.10% vs 24.20±2.19%, P<.01). Oocytes from Apc12+/- mice exhibited metaphase I arrest compared with oocytes from wild-type mice (72.29±0.51% vs 23.33±5.82%, P<.01), which could be rescued by injecting Apc12 cRNA (53.44±1.20%). CONCLUSION We identified a pathogenic mutation in APC12 in a female patient and confirmed its relevance as a causative factor for metaphase I arrest in oocytes, implying its importance as an APC/C component in the pathophysiology of oocyte maturation arrest, which caused female infertility.
Collapse
Affiliation(s)
- Yunying Lin
- Center for Reproductive Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China; Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai, China
| | - Zhe Wei
- Center for Reproductive Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China; Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai, China
| | - Ling Zhang
- Center for Reproductive Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China; Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai, China
| | - Yejie Yao
- Center for Reproductive Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China; Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai, China
| | - Yi Huang
- Center for Reproductive Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China; Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai, China
| | - Guangxin Yao
- Center for Reproductive Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China; Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai, China
| | - Wangsheng Wang
- Center for Reproductive Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China; Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai, China
| | - Shuanggang Hu
- Center for Reproductive Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China; Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai, China
| | - Ying Ding
- Center for Reproductive Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China; Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai, China
| | - Yao Lu
- Center for Reproductive Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China; Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai, China
| | - Xuejiao Bian
- Center for Reproductive Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China; Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai, China
| | - Xinyi Dong
- Center for Reproductive Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China; Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai, China
| | - Hengyu Guan
- Center for Reproductive Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China; Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai, China
| | - Yunfei Huang
- Center for Reproductive Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China; Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai, China
| | - Yun Sun
- Center for Reproductive Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China; Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai, China.
| |
Collapse
|
5
|
Zhang Z, Yu R, Shi Q, Wu ZJ, Li Q, Mu J, Chen B, Shi J, Ni R, Wu L, Li Q, Fu J, Li R, Sun X, Wang J, He L, Kuang Y, Sang Q, Wang L. COX15 deficiency causes oocyte ferroptosis. Proc Natl Acad Sci U S A 2024; 121:e2406174121. [PMID: 39471219 PMCID: PMC11551447 DOI: 10.1073/pnas.2406174121] [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: 03/26/2024] [Accepted: 09/19/2024] [Indexed: 11/01/2024] Open
Abstract
Mitochondria play diverse roles in mammalian physiology. The architecture, activity, and physiological functions of mitochondria in oocytes are largely different from those in somatic cells, but the mitochondrial proteins related to oocyte quality and reproductive longevity remain largely unknown. Here, using whole-exome sequencing data from 1,024 women (characterized by oocyte maturation arrest and degenerated or morphologically abnormal oocytes) and 2,868 healthy controls, we performed a population and gene-based burden test for mitochondrial genes and identified a candidate gene, cytochrome c oxidase assembly protein 15 (COX15). We report that biallelic COX15 pathogenic variants cause human oocyte ferroptosis and female infertility in a recessive inheritance pattern. COX15 variants impaired mitochondrial respiration in Saccharomyces cerevisiae and led to reduced protein levels in HeLa cells. Oocyte-specific deletion of Cox15 led to impaired Fe2+ and reactive oxygen species homeostasis that caused mitochondrial dysfunction and ultimately sensitized oocytes to ferroptosis. In addition, ferrostatin-1 (an inhibitor of ferroptosis) could rescue the oocyte ferroptosis phenotype in vitro and ex vivo. Our findings not only provide a genetic diagnostic marker for oocyte development defects but also expand the spectrum of mitochondrial disorders to female infertility and contribute to unique insights into the role of ferroptosis in human oocyte defects.
Collapse
Affiliation(s)
- Zhihua Zhang
- Institute of Pediatrics, Children’s Hospital of Fudan University, The Institutes of Biomedical Sciences, State Key Laboratory of Genetic Engineering, Fudan University, Shanghai200032, China
| | - Ran Yu
- Institute of Pediatrics, Children’s Hospital of Fudan University, The Institutes of Biomedical Sciences, State Key Laboratory of Genetic Engineering, Fudan University, Shanghai200032, China
| | - Qiuwen Shi
- Reproductive Medicine Center, The Third Affiliated Hospital, Guangxi Medical University, Nanning530031, Guangxi, China
| | - Zhi-Jing Wu
- The State Key Laboratory of Molecular Biology, Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai200031, China
- Key Laboratory of Systems Health Science of Zhejiang Province, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou310024, China
| | - Qingchun Li
- Department of Reproductive Medicine, Binzhou Medical University Hospital, Binzhou256603, China
| | - Jian Mu
- Institute of Pediatrics, Children’s Hospital of Fudan University, The Institutes of Biomedical Sciences, State Key Laboratory of Genetic Engineering, Fudan University, Shanghai200032, China
| | - Biaobang Chen
- Key Lab of Reproduction Regulation (Shanghai Institute for Biomedical and Pharmaceutical Technologies), National Health Commission of the People’s Republic of China, Shanghai200032, China
| | - Juanzi Shi
- Reproductive Medicine Center, Northwest Women’s and Children’s Hospital, Xi’an710069, China
| | - Renmin Ni
- Department of Reproductive Medicine, Kunming Angel Women’s and Children’s Hospital, Kunming650031, Yunnan, China
| | - Ling Wu
- Department of Assisted Reproduction, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai200011, China
| | - Qiaoli Li
- Institute of Pediatrics, Children’s Hospital of Fudan University, The Institutes of Biomedical Sciences, State Key Laboratory of Genetic Engineering, Fudan University, Shanghai200032, China
| | - Jing Fu
- Shanghai Ji Ai Genetics and In Vitro Fertilization Institute, Obstetrics and Gynecology Hospital, Fudan University, Shanghai200011, China
| | - Rong Li
- Reproductive Medicine Center, The Third Affiliated Hospital, Guangxi Medical University, Nanning530031, Guangxi, China
| | - Xiaoxi Sun
- Shanghai Ji Ai Genetics and In Vitro Fertilization Institute, Obstetrics and Gynecology Hospital, Fudan University, Shanghai200011, China
| | - Jiucun Wang
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Human Phenome Institute, Zhangjiang Fudan International Innovation Center, Fudan University, Shanghai200438, China
| | - Lin He
- Bio-X Center, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Shanghai Jiao Tong University, Shanghai200030, China
| | - Yanping Kuang
- Department of Assisted Reproduction, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai200011, China
| | - Qing Sang
- Institute of Pediatrics, Children’s Hospital of Fudan University, The Institutes of Biomedical Sciences, State Key Laboratory of Genetic Engineering, Fudan University, Shanghai200032, China
| | - Lei Wang
- Institute of Pediatrics, Children’s Hospital of Fudan University, The Institutes of Biomedical Sciences, State Key Laboratory of Genetic Engineering, Fudan University, Shanghai200032, China
| |
Collapse
|
6
|
Li Y, Wang Y, Tan YQ, Yue Q, Guo Y, Yan R, Meng L, Zhai H, Tong L, Yuan Z, Li W, Wang C, Han S, Ren S, Yan Y, Wang W, Gao L, Tan C, Hu T, Zhang H, Liu L, Yang P, Jiang W, Ye Y, Tan H, Wang Y, Lu C, Li X, Xie J, Yuan G, Cui Y, Shen B, Wang C, Guan Y, Li W, Shi Q, Lin G, Ni T, Sun Z, Ye L, Vourekas A, Guo X, Lin M, Zheng K. The landscape of RNA binding proteins in mammalian spermatogenesis. Science 2024; 386:eadj8172. [PMID: 39208083 DOI: 10.1126/science.adj8172] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Revised: 04/08/2024] [Accepted: 08/20/2024] [Indexed: 09/04/2024]
Abstract
Despite continuous expansion of the RNA binding protein (RBP) world, there is a lack of systematic understanding of RBPs in the mammalian testis, which harbors one of the most complex tissue transcriptomes. We adapted RNA interactome capture to mouse male germ cells, building an RBP atlas characterized by multiple layers of dynamics along spermatogenesis. Trapping of RNA-cross-linked peptides showed that the glutamic acid-arginine (ER) patch, a residue-coevolved polyampholytic element present in coiled coils, enhances RNA binding of its host RBPs. Deletion of this element in NONO (non-POU domain-containing octamer-binding protein) led to a defective mitosis-to-meiosis transition due to compromised NONO-RNA interactions. Whole-exome sequencing of over 1000 infertile men revealed a prominent role of RBPs in the human genetic architecture of male infertility and identified risk ER patch variants.
Collapse
Affiliation(s)
- Yang Li
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing 211166, China
| | - Yuanyuan Wang
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing 211166, China
- Department of Neurobiology, School of Basic Medical Science, Nanjing Medical University, Nanjing 211166, China
| | - Yue-Qiu Tan
- Institute of Reproductive and Stem Cell Engineering, NHC Key Laboratory of Human Stem Cell and Reproductive Engineering, School of Basic Medical Science, Central South University, Changsha 410083, China
- Clinical Research Center for Reproduction and Genetics in Hunan Province, Reproductive and Genetic Hospital of CITIC-Xiangya, Changsha 410008, China
| | - Qiuling Yue
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing 211166, China
- Department of Andrology, Nanjing Drum Tower Hospital, the Affiliated Hospital of Nanjing University, Nanjing 210008, China
| | - Yueshuai Guo
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing 211166, China
| | - Ruoyu Yan
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing 211166, China
- College of Life Sciences, Northwest A&F University, Yangling 712100, China
| | - Lanlan Meng
- Institute of Reproductive and Stem Cell Engineering, NHC Key Laboratory of Human Stem Cell and Reproductive Engineering, School of Basic Medical Science, Central South University, Changsha 410083, China
- Clinical Research Center for Reproduction and Genetics in Hunan Province, Reproductive and Genetic Hospital of CITIC-Xiangya, Changsha 410008, China
| | - Huicong Zhai
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing 211166, China
| | - Lingxiu Tong
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing 211166, China
| | - Zihan Yuan
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing 211166, China
| | - Wu Li
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing 211166, China
| | - Cuicui Wang
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing 211166, China
| | - Shenglin Han
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing 211166, China
| | - Sen Ren
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing 211166, China
| | - Yitong Yan
- Department of Neurobiology, School of Basic Medical Science, Nanjing Medical University, Nanjing 211166, China
| | - Weixu Wang
- Institute of Computational Biology, Helmholtz Center Munich, Munich 85764, Germany
| | - Lei Gao
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing 211166, China
| | - Chen Tan
- Institute of Reproductive and Stem Cell Engineering, NHC Key Laboratory of Human Stem Cell and Reproductive Engineering, School of Basic Medical Science, Central South University, Changsha 410083, China
| | - Tongyao Hu
- Institute of Reproductive and Stem Cell Engineering, NHC Key Laboratory of Human Stem Cell and Reproductive Engineering, School of Basic Medical Science, Central South University, Changsha 410083, China
| | - Hao Zhang
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing 211166, China
| | - Liya Liu
- Department of Neurobiology, School of Basic Medical Science, Nanjing Medical University, Nanjing 211166, China
| | - Pinglan Yang
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing 211166, China
| | - Wanyin Jiang
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing 211166, China
| | - Yiting Ye
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing 211166, China
| | - Huanhuan Tan
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing 211166, China
| | - Yanfeng Wang
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing 211166, China
| | - Chenyu Lu
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing 211166, China
| | - Xin Li
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing 211166, China
| | - Jie Xie
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing 211166, China
| | - Gege Yuan
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing 211166, China
| | - Yiqiang Cui
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing 211166, China
| | - Bin Shen
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing 211166, China
| | - Cheng Wang
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing 211166, China
- Department of Bioinformatics, School of Biomedical Engineering and Informatics, Nanjing Medical University, Nanjing 211166, China
| | - Yichun Guan
- Center for Reproductive Medicine, the Third Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Wei Li
- Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou 510623, China
| | - Qinghua Shi
- Division of Reproduction and Genetics, First Affiliated Hospital of USC, Hefei National Laboratory for Physical Sciences at Microscale, School of Basic Medical Sciences, Division of Life Sciences and Medicine, Biomedical Sciences and Health Laboratory of Anhui Province, University of Science and Technology of China, Hefei 230027, China
| | - Ge Lin
- Institute of Reproductive and Stem Cell Engineering, NHC Key Laboratory of Human Stem Cell and Reproductive Engineering, School of Basic Medical Science, Central South University, Changsha 410083, China
- Clinical Research Center for Reproduction and Genetics in Hunan Province, Reproductive and Genetic Hospital of CITIC-Xiangya, Changsha 410008, China
| | - Ting Ni
- State Key Laboratory of Genetic Engineering, Collaborative Innovation Center of Genetics and Development, Human Phenome Institute, Shanghai Engineering Research Center of Industrial Microorganisms, School of Life Sciences and Huashan Hospital, Fudan University, Shanghai 200438, China
| | - Zheng Sun
- Department of Medicine, Baylor College of Medicine, Houston, TX 77030, USA
| | - Lan Ye
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing 211166, China
| | - Anastasios Vourekas
- Department of Biological Sciences, Louisiana State University, Baton Rouge, LA 70803, USA
| | - Xuejiang Guo
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing 211166, China
| | - Mingyan Lin
- Department of Neurobiology, School of Basic Medical Science, Nanjing Medical University, Nanjing 211166, China
- Changzhou Medical Center, The Affiliated Changzhou Second People's Hospital of Nanjing Medical University, Changzhou 213000, China
- Division of Birth Cohort Study, Fujian Maternity and Child Health Hospital, Fuzhou 350014, China
| | - Ke Zheng
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing 211166, China
| |
Collapse
|
7
|
Hu H, Wan X, Zhang H, Sun J, Meng F, Zhang S, Gu Y, Gong F, Zhao H, Lin G, Zheng W. Biallelic variants in α-tubulin isotypes cause female infertility characterised as recurrent preimplantation embryo arrest. J Med Genet 2024; 61:1045-1052. [PMID: 39209701 DOI: 10.1136/jmg-2024-110163] [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: 06/04/2024] [Accepted: 08/22/2024] [Indexed: 09/04/2024]
Abstract
BACKGROUND Recurrent preimplantation embryo developmental arrest (RPEA) is the most common phenotype in assisted reproductive technology treatment failure associated with identified genetic abnormalities. Currently known maternal genetic variants explain only a limited number of cases. Variants of the β-tubulin subunit gene, TUBB8, cause oocyte meiotic arrest and RPEA through a broad spectrum of spindle defects. In contrast, α-tubulin subunit genes are poorly studied in the context of preimplantation embryonic development. METHODS Whole exome sequencing was performed on the PREA cohort. Functional characterisations of the identified candidate disease-causing variants were validated using Sanger sequencing, bioinformatics, in vitro functional analyses and single-cell RNA-sequencing of arrested embryos. RESULTS Four homozygous variants were identified in the PREA cohort: two of TUBA1C (p.Gln358Ter and p.Asp444Metfs*42) and two of TUBA4A (p.Arg339Cys and p.Tyr440Ter). These variants cause varying degrees of spindle assembly defects. Additionally, we characterised changes in the human arrested embryo transcriptome carrying TUBA4A variants, with a particular focus on spindle organisation, chromosome segregation and mRNA decay. CONCLUSION Our findings identified TUBA1C as a novel genetic marker and expanded the genetic and phenotypic spectrum of TUBA4A in female infertility and RPEA, which altogether highlighted the importance of α-tubulin isotypes in preimplantation embryonic development.
Collapse
Affiliation(s)
- Huiling Hu
- Center for Medical Genetics, School of Life Sciences, Central South University, Changsha, Hunan, China
- College of Life Science, Hunan Normal University, Changsha, Hunan, China
- Clinical Research Center for Reproduction and Genetics in Hunan Province, Reproductive and Genetic Hospital of CITIC-XIANGYA, Changsha, Hunan, China
| | - Xian Wan
- The Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Honghui Zhang
- Center for Reproductive Medicine, Cheeloo College of Medicine, Shandong University, Jinan, Shangdong, China
| | - Jiaqi Sun
- Center for Medical Genetics, School of Life Sciences, Central South University, Changsha, Hunan, China
| | - Fei Meng
- Clinical Research Center for Reproduction and Genetics in Hunan Province, Reproductive and Genetic Hospital of CITIC-XIANGYA, Changsha, Hunan, China
| | - Shuoping Zhang
- Clinical Research Center for Reproduction and Genetics in Hunan Province, Reproductive and Genetic Hospital of CITIC-XIANGYA, Changsha, Hunan, China
| | - Yifan Gu
- Clinical Research Center for Reproduction and Genetics in Hunan Province, Reproductive and Genetic Hospital of CITIC-XIANGYA, Changsha, Hunan, China
- NHC Key Laboratory of Human Stem Cell and Reproductive Engineering, School of Basic Medical Sciences, Central South University, Changsha, Hunan, China
| | - Fei Gong
- Clinical Research Center for Reproduction and Genetics in Hunan Province, Reproductive and Genetic Hospital of CITIC-XIANGYA, Changsha, Hunan, China
- NHC Key Laboratory of Human Stem Cell and Reproductive Engineering, School of Basic Medical Sciences, Central South University, Changsha, Hunan, China
| | - Han Zhao
- Center for Reproductive Medicine, Cheeloo College of Medicine, Shandong University, Jinan, Shangdong, China
| | - Ge Lin
- Clinical Research Center for Reproduction and Genetics in Hunan Province, Reproductive and Genetic Hospital of CITIC-XIANGYA, Changsha, Hunan, China
- NHC Key Laboratory of Human Stem Cell and Reproductive Engineering, School of Basic Medical Sciences, Central South University, Changsha, Hunan, China
| | - Wei Zheng
- Center for Medical Genetics, School of Life Sciences, Central South University, Changsha, Hunan, China
- Clinical Research Center for Reproduction and Genetics in Hunan Province, Reproductive and Genetic Hospital of CITIC-XIANGYA, Changsha, Hunan, China
| |
Collapse
|
8
|
Wang Y, Yang C, Sun H, Jiang H, Zhang P, Huang Y, Liu Z, Yu Y, Xu Z, Xiang H, Yi C. The Role of N6-methyladenosine Modification in Gametogenesis and Embryogenesis: Impact on Fertility. GENOMICS, PROTEOMICS & BIOINFORMATICS 2024; 22:qzae050. [PMID: 38937660 PMCID: PMC11514847 DOI: 10.1093/gpbjnl/qzae050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Revised: 06/02/2024] [Accepted: 06/24/2024] [Indexed: 06/29/2024]
Abstract
The most common epigenetic modification of messenger RNAs (mRNAs) is N6-methyladenosine (m6A), which is mainly located near the 3' untranslated region of mRNAs, near the stop codons, and within internal exons. The biological effect of m6A is dynamically modulated by methyltransferases (writers), demethylases (erasers), and m6A-binding proteins (readers). By controlling post-transcriptional gene expression, m6A has a significant impact on numerous biological functions, including RNA transcription, translation, splicing, transport, and degradation. Hence, m6A influences various physiological and pathological processes, such as spermatogenesis, oogenesis, embryogenesis, placental function, and human reproductive system diseases. During gametogenesis and embryogenesis, genetic material undergoes significant changes, including epigenomic modifications such as m6A. From spermatogenesis and oogenesis to the formation of an oosperm and early embryogenesis, m6A changes occur at every step. m6A abnormalities can lead to gamete abnormalities, developmental delays, impaired fertilization, and maternal-to-zygotic transition blockage. Both mice and humans with abnormal m6A modifications exhibit impaired fertility. In this review, we discuss the dynamic biological effects of m6A and its regulators on gamete and embryonic development and review the possible mechanisms of infertility caused by m6A changes. We also discuss the drugs currently used to manipulate m6A and provide prospects for the prevention and treatment of infertility at the epigenetic level.
Collapse
Affiliation(s)
- Yujie Wang
- Department of Obstetrics and Gynecology, the First Affiliated Hospital of Anhui Medical University, Hefei 230022, China
- NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract (Anhui Medical University), Hefei 230032, China
- MOE Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Hefei 230032, China
| | - Chen Yang
- Department of Obstetrics and Gynecology, the First Affiliated Hospital of Anhui Medical University, Hefei 230022, China
- NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract (Anhui Medical University), Hefei 230032, China
- MOE Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Hefei 230032, China
| | - Hanxiao Sun
- State Key Laboratory of Protein and Plant Gene Research, School of Life Sciences, Peking University, Beijing 100871, China
| | - Hui Jiang
- Department of Interventional Therapy, the Second Affiliated Hospital of Anhui Medical University, Hefei 230601, China
| | - Pin Zhang
- Department of Obstetrics and Gynecology, the First Affiliated Hospital of Anhui Medical University, Hefei 230022, China
- NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract (Anhui Medical University), Hefei 230032, China
- MOE Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Hefei 230032, China
| | - Yue Huang
- Department of Obstetrics and Gynecology, the First Affiliated Hospital of Anhui Medical University, Hefei 230022, China
- NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract (Anhui Medical University), Hefei 230032, China
- MOE Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Hefei 230032, China
| | - Zhenran Liu
- Department of Obstetrics and Gynecology, the First Affiliated Hospital of Anhui Medical University, Hefei 230022, China
- NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract (Anhui Medical University), Hefei 230032, China
- MOE Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Hefei 230032, China
| | - Yaru Yu
- Department of Obstetrics and Gynecology, the First Affiliated Hospital of Anhui Medical University, Hefei 230022, China
- NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract (Anhui Medical University), Hefei 230032, China
- MOE Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Hefei 230032, China
| | - Zuying Xu
- Department of Obstetrics and Gynecology, the First Affiliated Hospital of Anhui Medical University, Hefei 230022, China
- NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract (Anhui Medical University), Hefei 230032, China
- MOE Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Hefei 230032, China
| | - Huifen Xiang
- Department of Obstetrics and Gynecology, the First Affiliated Hospital of Anhui Medical University, Hefei 230022, China
- NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract (Anhui Medical University), Hefei 230032, China
- MOE Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Hefei 230032, China
| | - Chengqi Yi
- State Key Laboratory of Protein and Plant Gene Research, School of Life Sciences, Peking University, Beijing 100871, China
- Peking-Tsinghua Center for Life Sciences, Peking University, Beijing 100871, China
- Department of Chemical Biology and Synthetic and Functional Biomolecules Center, College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, China
| |
Collapse
|
9
|
Yang G, Wang Y, Hu S, Chen J, Chen L, Miao H, Li N, Luo H, He Y, Qian Y, Miao C, Feng R. Inhibition of neddylation disturbs zygotic genome activation through histone modification change and leads to early development arrest in mouse embryos. Biochim Biophys Acta Mol Basis Dis 2024; 1870:167292. [PMID: 38871031 DOI: 10.1016/j.bbadis.2024.167292] [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: 10/23/2023] [Revised: 05/09/2024] [Accepted: 06/06/2024] [Indexed: 06/15/2024]
Abstract
Post-translational modification and fine-tuned protein turnover are of great importance in mammalian early embryo development. Apart from the classic protein degradation promoting ubiquitination, new forms of ubiquitination-like modification are yet to be fully understood. Here, we demonstrate the function and potential mechanisms of one ubiquitination-like modification, neddylation, in mouse preimplantation embryo development. Treated with specific inhibitors, zygotes showed a dramatically decreased cleavage rate and almost all failed to enter the 4-cell stage. Transcriptional profiling showed genes were differentially expressed in pathways involving cell fate determination and cell differentiation, including several down-regulated zygotic genome activation (ZGA) marker genes. A decreased level of phosphorylated RNA polymerase II was detected, indicating impaired gene transcription inside the embryo cell nucleus. Proteomic data showed that differentially expressed proteins were enriched in histone modifications. We confirmed the lowered in methyltransferase (KMT2D) expression and a decrease in histone H3K4me3. At the same time, acetyltransferase (CBP/p300) reduced, while deacetylase (HDAC6) increased, resulting in an attenuation in histone H3K27ac. Additionally, we observed the up-regulation in YAP1 and RPL13 activities, indicating potential abnormalities in the downstream response of Hippo signaling pathway. In summary, we found that inhibition of neddylation induced epigenetic changes in early embryos and led to abnormalities in related downstream signaling pathways. This study sheds light upon new forms of ubiquitination regulating mammalian embryonic development and may contribute to further investigation of female infertility pathology.
Collapse
Affiliation(s)
- Guangping Yang
- State Key Laboratory of Reproduction Medicine and Offspring Health, Nanjing Medical University, Nanjing, Jiangsu 210029, China; Yangzhou Maternal and Child Health Care Hospital Affiliated to Yangzhou University, China
| | - Yingnan Wang
- State Key Laboratory of Reproduction Medicine and Offspring Health, Nanjing Medical University, Nanjing, Jiangsu 210029, China
| | - Saifei Hu
- State Key Laboratory of Reproduction Medicine and Offspring Health, Nanjing Medical University, Nanjing, Jiangsu 210029, China
| | - Jianhua Chen
- State Key Laboratory of Reproduction Medicine and Offspring Health, Nanjing Medical University, Nanjing, Jiangsu 210029, China
| | - Liangliang Chen
- State Key Laboratory of Reproduction Medicine and Offspring Health, Nanjing Medical University, Nanjing, Jiangsu 210029, China
| | - Hui Miao
- Department of Reproductive Genetics, Heping Hospital of Changzhi Medical College, Key Laboratory of Reproduction Engineer of Shanxi Health Committee, Changzhi, Shanxi 046000, China
| | - Na Li
- Department of Reproductive Genetics, Heping Hospital of Changzhi Medical College, Key Laboratory of Reproduction Engineer of Shanxi Health Committee, Changzhi, Shanxi 046000, China
| | - Hui Luo
- State Key Laboratory of Reproduction Medicine and Offspring Health, Nanjing Medical University, Nanjing, Jiangsu 210029, China
| | - Yanni He
- State Key Laboratory of Reproduction Medicine and Offspring Health, Nanjing Medical University, Nanjing, Jiangsu 210029, China
| | - Yun Qian
- Clinical Center of Reproductive Medicine, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210008, China
| | - Congxiu Miao
- Department of Reproductive Genetics, Heping Hospital of Changzhi Medical College, Key Laboratory of Reproduction Engineer of Shanxi Health Committee, Changzhi, Shanxi 046000, China.
| | - Ruizhi Feng
- State Key Laboratory of Reproduction Medicine and Offspring Health, Nanjing Medical University, Nanjing, Jiangsu 210029, China; Clinical Center of Reproductive Medicine, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210008, China; Innovation Center of Suzhou Nanjing Medical University, Suzhou, Jiangsu 215005, China.
| |
Collapse
|
10
|
Chachlaki K, Duc KL, Storme L, Prévot V. Novel insights into minipuberty and GnRH: Implications on neurodevelopment, cognition, and COVID-19 therapeutics. J Neuroendocrinol 2024; 36:e13387. [PMID: 38565500 PMCID: PMC7616535 DOI: 10.1111/jne.13387] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Revised: 03/18/2024] [Accepted: 03/18/2024] [Indexed: 04/04/2024]
Abstract
In humans, the first 1000 days of life are pivotal for brain and organism development. Shortly after birth, gonadotropin-releasing hormone (GnRH) neurons in the hypothalamus are activated, a phenomenon known as minipuberty. This phenomenon, observed in all mammals studied, influences the postnatal development of the hypothalamic-pituitary-gonadal (HPG) axis and reproductive function. This review will put into perspective the results of recent studies showing that the impact of minipuberty extends beyond reproductive function, influencing sensory and cognitive maturation. Studies in mice have revealed the role of nitric oxide (NO) in regulating minipuberty amplitude, with NO deficiency linked to cognitive and olfactory deficits. Additionally, findings indicate that cognitive and sensory defects in adulthood in a mouse model of Down syndrome are associated with an age-dependent decline of GnRH production, whose origin can be traced back to minipuberty, and point to the potential therapeutic role of pulsatile GnRH administration in cognitive disorders. Furthermore, this review delves into the repercussions of COVID-19 on GnRH production, emphasizing potential consequences for neurodevelopment and cognitive function in infected individuals. Notably, GnRH neurons appear susceptible to SARS-CoV-2 infection, raising concerns about potential long-term effects on brain development and function. In conclusion, the intricate interplay between GnRH neurons, GnRH release, and the activity of various extrahypothalamic brain circuits reveals an unexpected role for these neuroendocrine neurons in the development and maintenance of sensory and cognitive functions, supplementing their established function in reproduction. Therapeutic interventions targeting the HPG axis, such as inhaled NO therapy in infancy and pulsatile GnRH administration in adults, emerge as promising approaches for addressing neurodevelopmental cognitive disorders and pathological aging.
Collapse
Affiliation(s)
- Konstantina Chachlaki
- Univ. Lille, Inserm, CHU Lille, Laboratory of Development and Plasticity of the Neuroendocrine Brain, Lille Neuroscience & Cognition, UMR_S1172, Lille, France
- Univ. Lille, Inserm, CHU Lille, Hospital-University Federation (FHU) 1000 first days of Life, Lille, France
| | - Kevin Le Duc
- Univ. Lille, Inserm, CHU Lille, Hospital-University Federation (FHU) 1000 first days of Life, Lille, France
- CHU Lille, Neonatology Department, Jeanne de Flandres Hospital, Lille, France
| | - Laurent Storme
- Univ. Lille, Inserm, CHU Lille, Hospital-University Federation (FHU) 1000 first days of Life, Lille, France
- CHU Lille, Neonatology Department, Jeanne de Flandres Hospital, Lille, France
| | - Vincent Prévot
- Univ. Lille, Inserm, CHU Lille, Laboratory of Development and Plasticity of the Neuroendocrine Brain, Lille Neuroscience & Cognition, UMR_S1172, Lille, France
- Univ. Lille, Inserm, CHU Lille, Hospital-University Federation (FHU) 1000 first days of Life, Lille, France
| |
Collapse
|
11
|
Chen S, Fan Y, Sun Y, Li S, Zheng Z, Chu C, Li L, Yin C. Identification and functional characteristics of CHD1L gene variants implicated in human Müllerian duct anomalies. Biol Res 2024; 57:68. [PMID: 39342328 PMCID: PMC11437902 DOI: 10.1186/s40659-024-00550-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: 06/11/2024] [Accepted: 09/16/2024] [Indexed: 10/01/2024] Open
Abstract
BACKGROUND Müllerian duct anomalies (MDAs) are congenital developmental disorders that present as a series of abnormalities within the reproductive tracts of females. Genetic factors are linked to MDAs and recent advancements in whole-exome sequencing (WES) provide innovative perspectives in this field. However, relevant mechanism has only been investigated in a restricted manner without clear elucidation of respective observations. METHODS Our previous study reported that 2 of 12 patients with MDAs harbored the CHD1L variant c.348-1G>C. Subsequently, an additional 85 MDAs patients were recruited. Variants in CHD1L were screened through the in-house database of WES performed in the cohort and two cases were identified. One presented with partial septate uterus with left renal agenesis and the other with complete septate uterus, duplicated cervices and longitudinal vaginal septum. The pathogenicity of the discovered variants was further assessed by molecular dynamics simulation and various functional assays. RESULTS Ultimately, two novel heterozygous CHD1L variants, including a missense variant c.956G>A (p.R319Q) and a nonsense variant c.1831C>T (p.R611*) were observed. The variants were absent in 100 controls. Altogether, the contribution yield of CHD1L to MDAs was calculated as 4.12% (4/97). All three variants were assessed as pathogenic through various functional analysis. The splice-site variant c.348-1G>C resulted in a 11 bp sequence skipping in exon 4 of CHD1L and led to nonsense mediated decay of its transcripts. Unlike WT CHD1L, the truncated R611* protein mislocalized to the cytoplasm, abolish the ability of CHD1L to promote cell migration and failed to interact with PARP1 owing to the loss of macro domain. The R319Q variant exhibited conformational disparities and showed abnormal protein recruitment behavior through laser microirradiation comparing with the WT CHD1L. All these variants impaired the CHD1L function in DNA damage repair, thus participating in MDAs. CONCLUSIONS The current study not only expands the mutational spectrum of CHD1L in MDAs but determines three variants as pathogenic according to ACMG guidelines with reliable functional evidence. Additionally, the impairment in DNA damage repair is an underlying mechanism involved in MDAs.
Collapse
Affiliation(s)
- Shuya Chen
- Central Laboratory, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, 17 QiHeLou Street, Dongcheng District, Beijing, 100006, China
| | - Yali Fan
- Central Laboratory, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, 17 QiHeLou Street, Dongcheng District, Beijing, 100006, China
| | - Yujun Sun
- Central Laboratory, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, 17 QiHeLou Street, Dongcheng District, Beijing, 100006, China
| | - Shenghui Li
- Department of Gynecology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, 251 YaoJiaYuan Road, Chaoyang District, Beijing, 100026, China
| | - Zhi Zheng
- Central Laboratory, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, 17 QiHeLou Street, Dongcheng District, Beijing, 100006, China
| | - Chunfang Chu
- Department of Gynecology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, 251 YaoJiaYuan Road, Chaoyang District, Beijing, 100026, China.
| | - Lin Li
- Central Laboratory, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, 17 QiHeLou Street, Dongcheng District, Beijing, 100006, China.
| | - Chenghong Yin
- Central Laboratory, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, 17 QiHeLou Street, Dongcheng District, Beijing, 100006, China.
| |
Collapse
|
12
|
Ye JW, Abbas T, Zhou JT, Chen J, Yang ML, Huang XH, Zhang H, Ma H, Ma A, Xu B, Murtaza G, Shi QH, Shi BL. Homozygous CCDC146 mutation causes oligoasthenoteratozoospermia in humans and mice. Zool Res 2024; 45:1073-1087. [PMID: 39245651 PMCID: PMC11491774 DOI: 10.24272/j.issn.2095-8137.2024.019] [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: 05/11/2024] [Accepted: 06/11/2024] [Indexed: 09/10/2024] Open
Abstract
Infertility represents a significant health concern, with sperm quantity and quality being crucial determinants of male fertility. Oligoasthenoteratozoospermia (OAT) is characterized by reduced sperm motility, lower sperm concentration, and morphological abnormalities in sperm heads and flagella. Although variants in several genes have been implicated in OAT, its genetic etiologies and pathogenetic mechanisms remain inadequately understood. In this study, we identified a homozygous nonsense mutation (c.916C>T, p.Arg306*) in the coiled-coil domain containing 146 ( CCDC146) gene in an infertile male patient with OAT. This mutation resulted in the production of a truncated CCDC146 protein (amino acids 1-305), retaining only two out of five coiled-coil domains. To validate the pathogenicity of the CCDC146 mutation, we generated a mouse model ( Ccdc146 mut/mut ) with a similar mutation to that of the patient. Consistently, the Ccdc146 mut/mut mice exhibited infertility, characterized by significantly reduced sperm counts, diminished motility, and multiple defects in sperm heads and flagella. Furthermore, the levels of axonemal proteins, including DNAH17, DNAH1, and SPAG6, were significantly reduced in the sperm of Ccdc146 mut/mut mice. Additionally, both human and mouse CCDC146 interacted with intraflagellar transport protein 20 (IFT20), but this interaction was lost in the mutated versions, leading to the degradation of IFT20. This study identified a novel deleterious homozygous nonsense mutation in CCDC146 that causes male infertility, potentially by disrupting axonemal protein transportation. These findings offer valuable insights for genetic counseling and understanding the mechanisms underlying CCDC146 mutant-associated infertility in human males.
Collapse
Affiliation(s)
- Jing-Wei Ye
- Division of Reproduction and Genetics, First Affiliated Hospital of University of Science and Technology of China, Hefei National Laboratory for Physical Sciences at Microscale, School of Basic Medical Sciences, Division of Life Sciences and Medicine, Biomedical Sciences and Health Laboratory of Anhui Province, Institute of Health and Medicine, Hefei Comprehensive National Science Center, University of Science and Technology of China, Hefei, Anhui 230026, China
| | - Tanveer Abbas
- Division of Reproduction and Genetics, First Affiliated Hospital of University of Science and Technology of China, Hefei National Laboratory for Physical Sciences at Microscale, School of Basic Medical Sciences, Division of Life Sciences and Medicine, Biomedical Sciences and Health Laboratory of Anhui Province, Institute of Health and Medicine, Hefei Comprehensive National Science Center, University of Science and Technology of China, Hefei, Anhui 230026, China
| | - Jian-Teng Zhou
- Division of Reproduction and Genetics, First Affiliated Hospital of University of Science and Technology of China, Hefei National Laboratory for Physical Sciences at Microscale, School of Basic Medical Sciences, Division of Life Sciences and Medicine, Biomedical Sciences and Health Laboratory of Anhui Province, Institute of Health and Medicine, Hefei Comprehensive National Science Center, University of Science and Technology of China, Hefei, Anhui 230026, China
| | - Jing Chen
- Division of Reproduction and Genetics, First Affiliated Hospital of University of Science and Technology of China, Hefei National Laboratory for Physical Sciences at Microscale, School of Basic Medical Sciences, Division of Life Sciences and Medicine, Biomedical Sciences and Health Laboratory of Anhui Province, Institute of Health and Medicine, Hefei Comprehensive National Science Center, University of Science and Technology of China, Hefei, Anhui 230026, China
| | - Meng-Lei Yang
- Division of Reproduction and Genetics, First Affiliated Hospital of University of Science and Technology of China, Hefei National Laboratory for Physical Sciences at Microscale, School of Basic Medical Sciences, Division of Life Sciences and Medicine, Biomedical Sciences and Health Laboratory of Anhui Province, Institute of Health and Medicine, Hefei Comprehensive National Science Center, University of Science and Technology of China, Hefei, Anhui 230026, China
| | - Xiong-Heng Huang
- Division of Reproduction and Genetics, First Affiliated Hospital of University of Science and Technology of China, Hefei National Laboratory for Physical Sciences at Microscale, School of Basic Medical Sciences, Division of Life Sciences and Medicine, Biomedical Sciences and Health Laboratory of Anhui Province, Institute of Health and Medicine, Hefei Comprehensive National Science Center, University of Science and Technology of China, Hefei, Anhui 230026, China
| | - Huan Zhang
- Division of Reproduction and Genetics, First Affiliated Hospital of University of Science and Technology of China, Hefei National Laboratory for Physical Sciences at Microscale, School of Basic Medical Sciences, Division of Life Sciences and Medicine, Biomedical Sciences and Health Laboratory of Anhui Province, Institute of Health and Medicine, Hefei Comprehensive National Science Center, University of Science and Technology of China, Hefei, Anhui 230026, China
| | - Hui Ma
- Division of Reproduction and Genetics, First Affiliated Hospital of University of Science and Technology of China, Hefei National Laboratory for Physical Sciences at Microscale, School of Basic Medical Sciences, Division of Life Sciences and Medicine, Biomedical Sciences and Health Laboratory of Anhui Province, Institute of Health and Medicine, Hefei Comprehensive National Science Center, University of Science and Technology of China, Hefei, Anhui 230026, China
| | - Ao Ma
- Division of Reproduction and Genetics, First Affiliated Hospital of University of Science and Technology of China, Hefei National Laboratory for Physical Sciences at Microscale, School of Basic Medical Sciences, Division of Life Sciences and Medicine, Biomedical Sciences and Health Laboratory of Anhui Province, Institute of Health and Medicine, Hefei Comprehensive National Science Center, University of Science and Technology of China, Hefei, Anhui 230026, China
- Institute of Andrology, Nanjing Drum Tower Hospital, Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu 210008, China
| | - Bo Xu
- Division of Reproduction and Genetics, First Affiliated Hospital of University of Science and Technology of China, Hefei National Laboratory for Physical Sciences at Microscale, School of Basic Medical Sciences, Division of Life Sciences and Medicine, Biomedical Sciences and Health Laboratory of Anhui Province, Institute of Health and Medicine, Hefei Comprehensive National Science Center, University of Science and Technology of China, Hefei, Anhui 230026, China
| | - Ghulam Murtaza
- Division of Reproduction and Genetics, First Affiliated Hospital of University of Science and Technology of China, Hefei National Laboratory for Physical Sciences at Microscale, School of Basic Medical Sciences, Division of Life Sciences and Medicine, Biomedical Sciences and Health Laboratory of Anhui Province, Institute of Health and Medicine, Hefei Comprehensive National Science Center, University of Science and Technology of China, Hefei, Anhui 230026, China
| | - Qing-Hua Shi
- Division of Reproduction and Genetics, First Affiliated Hospital of University of Science and Technology of China, Hefei National Laboratory for Physical Sciences at Microscale, School of Basic Medical Sciences, Division of Life Sciences and Medicine, Biomedical Sciences and Health Laboratory of Anhui Province, Institute of Health and Medicine, Hefei Comprehensive National Science Center, University of Science and Technology of China, Hefei, Anhui 230026, China. E-mail:
| | - Bao-Lu Shi
- Division of Reproduction and Genetics, First Affiliated Hospital of University of Science and Technology of China, Hefei National Laboratory for Physical Sciences at Microscale, School of Basic Medical Sciences, Division of Life Sciences and Medicine, Biomedical Sciences and Health Laboratory of Anhui Province, Institute of Health and Medicine, Hefei Comprehensive National Science Center, University of Science and Technology of China, Hefei, Anhui 230026, China. E-mail:
| |
Collapse
|
13
|
Pan M, Qi Q, Li C, Wang J, Pan X, Zhou J, Sun H, Li L, Wang L. Effect and mechanism of Hashimoto thyroiditis on female infertility: A clinical trial, bioinformatics analysis, and experiments-based study. Biosci Trends 2024; 18:356-369. [PMID: 38925961 DOI: 10.5582/bst.2024.01120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/28/2024]
Abstract
Diagnosing Hashimoto thyroiditis (HT) relies on thyroglobulin antibody (TgAb) and thyroid peroxidase antibody (TPOAb) titers. The influence of these antibodies on female infertility remains a subject of debate. This study aims to explore the effect and mechanism of HT on female infertility. First, a single-center cross-sectional study was conducted to investigate whether TgAb and TPOAb are the key factors leading to female infertility. Second, bioinformatic analysis was performed to investigate the potential target molecules and pathways. Third, in vivo experiments were performed to explore the effects of elevated TgAb levels on embryo implantation in a mouse model of autoimmune thyroiditis (AIT). Four hundred and five infertile women and 155 healthy controls were enrolled in the cross-sectional study. Results indicated that the TPOAb titer was associated with female infertility, while the TgAb titer showed no significant association. The increased levels of TgAb and TPOAb are not significantly correlated with anti-Mullerian hormone. Bioinformatic analysis indicated that the common target molecules for HT and female infertility include interleukin (IL)-6, IL-10, matrix metalloproteinase 9, and tumor necrosis factor, suggesting potential regulation through multiple signaling pathways such as HIF-1, VEGF, MAPK, and Th17 cell differentiation. A certain dose of porcine thyroglobulin can successfully establish a mouse model of AIT. In this mouse model, embryo implantation and ovarian reserve remain unaffected by elevated TgAb levels. In conclusion, the serum TPOAb titer was associated with infertility due to female factors but the TgAb titer showed no significant association. A simple increase in serum TgAb titer does not affect embryo implantation and ovarian reserve in the AIT model.
Collapse
Affiliation(s)
- Meijun Pan
- Laboratory for Reproductive Immunology, Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China
- The Academy of Integrative Medicine of Fudan University, Shanghai, China
- Shanghai Key Laboratory of Female Reproductive Endocrine-related Diseases, Shanghai, China
- The Second Clinical Medical College of Guangzhou University of Traditional Chinese Medicine, Guangzhou, China
| | - Qing Qi
- Wuhan Business University, Wuhan, Hubei, China
| | - Chuyu Li
- Laboratory for Reproductive Immunology, Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China
- The Academy of Integrative Medicine of Fudan University, Shanghai, China
- Shanghai Key Laboratory of Female Reproductive Endocrine-related Diseases, Shanghai, China
| | - Jing Wang
- Laboratory for Reproductive Immunology, Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China
- The Academy of Integrative Medicine of Fudan University, Shanghai, China
- Shanghai Key Laboratory of Female Reproductive Endocrine-related Diseases, Shanghai, China
| | - Xinyao Pan
- Laboratory for Reproductive Immunology, Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China
- The Academy of Integrative Medicine of Fudan University, Shanghai, China
- Shanghai Key Laboratory of Female Reproductive Endocrine-related Diseases, Shanghai, China
| | - Jing Zhou
- Department of Obstetrics and Gynecology, Nanfang Hospital of Southern Medical University, Guangzhou, China
| | - Hongmei Sun
- Laboratory for Reproductive Immunology, Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China
- The Academy of Integrative Medicine of Fudan University, Shanghai, China
- Shanghai Key Laboratory of Female Reproductive Endocrine-related Diseases, Shanghai, China
| | - Lisha Li
- Laboratory for Reproductive Immunology, Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China
- The Academy of Integrative Medicine of Fudan University, Shanghai, China
- Shanghai Key Laboratory of Female Reproductive Endocrine-related Diseases, Shanghai, China
| | - Ling Wang
- Laboratory for Reproductive Immunology, Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China
- The Academy of Integrative Medicine of Fudan University, Shanghai, China
- Shanghai Key Laboratory of Female Reproductive Endocrine-related Diseases, Shanghai, China
| |
Collapse
|
14
|
Xu Z, Lv C, Gao J, Cui Y, Liu W, He Z, He L. LncRNA ACVR2B-as1 interacts with ALDOA to regulate the self-renewal and apoptosis of human spermatogonial stem cells by controlling glycolysis activity. Cell Mol Life Sci 2024; 81:391. [PMID: 39254854 PMCID: PMC11387566 DOI: 10.1007/s00018-024-05414-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: 04/24/2024] [Revised: 08/03/2024] [Accepted: 08/14/2024] [Indexed: 09/11/2024]
Abstract
Human spermatogonial stem cells (SSCs) have significant applications in reproductive medicine and regenerative medicine because of their great plasticity. Nevertheless, it remains unknown about the functions and mechanisms of long non-coding RNA (LncRNA) in regulating the fate determinations of human SSCs. Here we have demonstrated that LncRNA ACVR2B-as1 (activin A receptor type 2B antisense RNA 1) controls the self-renewal and apoptosis of human SSCs by interaction with ALDOA via glycolysis activity. LncRNA ACVR2B-as1 is highly expressed in human SSCs. LncRNA ACVR2B-as1 silencing suppresses the proliferation and DNA synthesis and enhances the apoptosis of human SSCs. Mechanistically, our ChIRP-MS and RIP assays revealed that ACVR2B-as1 interacted with ALDOA in human SSCs. High expression of ACVR2B-as1 enhanced the proliferation, DNA synthesis, and glycolysis of human SSCs but inhibited their apoptosis through up-regulation of ALDOA. Importantly, overexpression of ALDOA counteracted the effect of ACVR2B-as1 knockdown on the aforementioned biological processes. Collectively, these results indicate that ACVR2B-as1 interacts with ALDOA to control the self-renewal and apoptosis of human SSCs by enhancing glycolysis activity. This study is of great significance because it sheds a novel insight into molecular mechanisms underlying the fate decisions of human SSCs and it may offer innovative approaches to address the etiology of male infertility.
Collapse
Affiliation(s)
- Zhipeng Xu
- Department of Urology, The Third Xiangya Hospital, Central South University, Changsha, 410011, China
| | - Cai Lv
- Department of Urology, Haikou Municipal Hospital, Haikou, Hainan, China
| | - Jun Gao
- Department of Urology, The Third Xiangya Hospital, Central South University, Changsha, 410011, China
| | - Yinghong Cui
- Key Laboratory of Model Animals and Stem Cell Biology in Hunan Province, Engineering Research Center of Reproduction and Translational Medicine of Hunan Province, Hunan Normal University School of Medicine, Changsha, 410013, Hunan, China
| | - Wei Liu
- Key Laboratory of Model Animals and Stem Cell Biology in Hunan Province, Engineering Research Center of Reproduction and Translational Medicine of Hunan Province, Hunan Normal University School of Medicine, Changsha, 410013, Hunan, China
- Department of Urology, Haikou Municipal Hospital, Haikou, Hainan, China
| | - Zuping He
- Key Laboratory of Model Animals and Stem Cell Biology in Hunan Province, Engineering Research Center of Reproduction and Translational Medicine of Hunan Province, Hunan Normal University School of Medicine, Changsha, 410013, Hunan, China.
- Shanghai Key Laboratory of Reproductive Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| | - Leye He
- Department of Urology, The Third Xiangya Hospital, Central South University, Changsha, 410011, China.
- Department of Pharmacy, The Third Xiangya Hospital, Central South University, Changsha, 410011, China.
| |
Collapse
|
15
|
Li X, Zou Y, Geng B, Liu P, Cao L, Zhang Z, Hu S, Wang C, Zhao Y, Wu Q, Tan J. Transcriptome analysis reveals that defects in cell cycle regulation contribute to preimplantation embryo arrest. Genomics 2024; 116:110946. [PMID: 39326642 DOI: 10.1016/j.ygeno.2024.110946] [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: 06/18/2024] [Revised: 09/09/2024] [Accepted: 09/23/2024] [Indexed: 09/28/2024]
Abstract
Patients with preimplantation embryo arrest (PREMBA) often experience assisted reproductive failure primarily due to the lack of transferable embryos, and the molecular mechanisms underlying PREMBA remain unclear. In our study, the embryos from five women with recurrent preimplantation embryo arrest and three women with tubal factor infertility were used for single-embryo transcriptome sequencing. Meanwhile, the transcriptomes of normal human preimplantation embryos obtained from GSE36552 were utilized to perform a comparative analysis with the transcriptomes of PREMBA embryos. Our results showed dysregulation of the cell cycle phase transition might be a potential pathogenic factor contributing to PREMBA. Through integrated analysis of the differentially expressed genes (DEGs) and weighted gene co-expression network analysis (WGCNA), we identified a number of hub genes using the protein-protein interaction network. The top 5 hub genes were as follows: CCNB2, BUB1B, CDC25A, CCNB3, and PLK3. The expression of hub genes was validated in PREMBA embryos and donated embryos using RT-qPCR. The knockdown of Ccnb2 in mouse zygotes led to an increase in embryo fragmentation, a rise in apoptosis, and a reduction in blastocyst formation. Furthermore, silencing the expression of CDC25A in HEK293T cells resulted in a decrease in cell proliferation and an increase in apoptosis, providing further support for our findings. Our findings could predict the development outcomes of preimplantation embryos and be used as potential therapeutic targets to prevent recurrent failures of IVF/ICSI attempts.
Collapse
Affiliation(s)
- Xin Li
- Reproductive Medicine Center, Jiangxi Maternal and Child Health Hospital, Nanchang, Jiangxi Province, China; JXHC Key Laboratory of Fertility Preservation, Jiangxi Maternal and Child Health Hospital, Nanchang, Jiangxi Province, China
| | - Yang Zou
- Central Laboratory, Jiangxi Maternal and Child Health Hospital, Nanchang, Jiangxi Province, China
| | - Baobao Geng
- Suzhou Institute of Systems Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, SuZhou, Jiangsu Province, China
| | - Peipei Liu
- Reproductive Medicine Center, Jiangxi Maternal and Child Health Hospital, Nanchang, Jiangxi Province, China; JXHC Key Laboratory of Fertility Preservation, Jiangxi Maternal and Child Health Hospital, Nanchang, Jiangxi Province, China
| | - Liyun Cao
- Reproductive Medicine Center, Jiangxi Maternal and Child Health Hospital, Nanchang, Jiangxi Province, China; JXHC Key Laboratory of Fertility Preservation, Jiangxi Maternal and Child Health Hospital, Nanchang, Jiangxi Province, China
| | - Zhiqin Zhang
- Reproductive Medicine Center, Jiangxi Maternal and Child Health Hospital, Nanchang, Jiangxi Province, China
| | - Shaofeng Hu
- JXHC Key Laboratory of Fertility Preservation, Jiangxi Maternal and Child Health Hospital, Nanchang, Jiangxi Province, China
| | - Changhua Wang
- JXHC Key Laboratory of Fertility Preservation, Jiangxi Maternal and Child Health Hospital, Nanchang, Jiangxi Province, China
| | - Yan Zhao
- Reproductive Medicine Center, Jiangxi Maternal and Child Health Hospital, Nanchang, Jiangxi Province, China.
| | - Qiongfang Wu
- Reproductive Medicine Center, Jiangxi Maternal and Child Health Hospital, Nanchang, Jiangxi Province, China.
| | - Jun Tan
- Reproductive Medicine Center, Jiangxi Maternal and Child Health Hospital, Nanchang, Jiangxi Province, China; JXHC Key Laboratory of Fertility Preservation, Jiangxi Maternal and Child Health Hospital, Nanchang, Jiangxi Province, China.
| |
Collapse
|
16
|
Wang H, Huang Z, Shen X, Lee Y, Song X, Shu C, Wu LH, Pakkiri LS, Lim PL, Zhang X, Drum CL, Zhu J, Li R. Rejuvenation of aged oocyte through exposure to young follicular microenvironment. NATURE AGING 2024; 4:1194-1210. [PMID: 39251866 DOI: 10.1038/s43587-024-00697-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Accepted: 07/30/2024] [Indexed: 09/11/2024]
Abstract
Reproductive aging is a major cause of fertility decline, attributed to decreased oocyte quantity and developmental potential. A possible cause is aging of the surrounding follicular somatic cells that support oocyte growth and development by providing nutrients and regulatory factors. Here, by creating chimeric follicles, whereby an oocyte from one follicle was transplanted into and cultured within another follicle whose native oocyte was removed, we show that young oocytes cultured in aged follicles exhibited impeded meiotic maturation and developmental potential, whereas aged oocytes cultured within young follicles were significantly improved in rates of maturation, blastocyst formation and live birth after in vitro fertilization and embryo implantation. This rejuvenation of aged oocytes was associated with enhanced interaction with somatic cells, transcriptomic and metabolomic remodeling, improved mitochondrial function and higher fidelity of meiotic chromosome segregation. These findings provide the basis for a future follicular somatic cell-based therapy to treat female infertility.
Collapse
Affiliation(s)
- HaiYang Wang
- Mechanobiology Institute, National University of Singapore, Singapore, Singapore.
| | - Zhongwei Huang
- NUS Bia Echo Asia Centre for Reproductive Longevity and Equality, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Department of Obstetrics and Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Xingyu Shen
- Mechanobiology Institute, National University of Singapore, Singapore, Singapore
| | - Yaelim Lee
- Mechanobiology Institute, National University of Singapore, Singapore, Singapore
| | - XinJie Song
- Mechanobiology Institute, National University of Singapore, Singapore, Singapore
| | - Chang Shu
- Mechanobiology Institute, National University of Singapore, Singapore, Singapore
| | - Lik Hang Wu
- Cardiovascular Research Institute, National University Health System, Singapore, Singapore
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Department of Pharmacy, Faculty of Science, National University of Singapore, Singapore, Singapore
| | - Leroy Sivappiragasam Pakkiri
- Cardiovascular Research Institute, National University Health System, Singapore, Singapore
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Poh Leong Lim
- Cardiovascular Research Institute, National University Health System, Singapore, Singapore
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Xi Zhang
- Center for Cell Dynamics and Department of Cell Biology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Chester Lee Drum
- Cardiovascular Research Institute, National University Health System, Singapore, Singapore
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Jin Zhu
- Mechanobiology Institute, National University of Singapore, Singapore, Singapore
| | - Rong Li
- Mechanobiology Institute, National University of Singapore, Singapore, Singapore.
- Center for Cell Dynamics and Department of Cell Biology, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
- Department of Biological Sciences, National University of Singapore, Singapore, Singapore.
| |
Collapse
|
17
|
Wang Y, Dong S, Li H, Yang Y, Guo AL, Chao L. Nomogram for predicting live birth in ovulatory women undergoing frozen-thawed embryo transfer. BMC Pregnancy Childbirth 2024; 24:559. [PMID: 39192200 DOI: 10.1186/s12884-024-06759-7] [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: 11/16/2023] [Accepted: 08/16/2024] [Indexed: 08/29/2024] Open
Abstract
BACKGROUND Study objectives included the development of a practical nomogram for predicting live birth following frozen-thawed embryo transfers in ovulatory women. METHODS Totally, 2884 patients with regular menstrual cycles in our center were retrospectively enrolled. In an 8:2 ratio, we randomly assigned patients to training and validation cohorts. Then we identified risk factors by multivariate logistic regression and constructed nomogram. Finally, receiver operating characteristic curve analysis, calibration curve and decision curve analysis were performed to assess the calibration and discriminative ability of the nomogram. RESULTS We identified five variables which were related to live birth, including age, anti-Müllerian hormone (AMH), protocol of frozen-thawed embryo transfer (FET), stage of embryos and amount of high-quality embryos. We then constructed nomograms that predict the probabilities of live birth by using those five parameters. Receiver operating characteristic curve analysis (ROC) showed that the area under the curve (AUC) for live birth was 0.666 (95% CI: 0.644-0.688) in the training cohort. The AUC in the subsequent validation cohorts was 0.669 (95% CI, 0.625-0.713). The clinical practicability of this nomogram was demonstrated through calibration curve analysis and decision curve analysis. CONCLUSIONS Our nomogram provides a visual and simple tool in predicting live birth in ovulatory women who received FET. It could also provide advice and guidance for physicians and patients on decision-making during the FET procedure.
Collapse
Affiliation(s)
- Ying Wang
- Department of Obstetrics and Gynecology, Center for Reproductive Medicine, Qilu Hospital, Shandong University, Ji'nan, Shandong, 250012, P.R. China
| | - Shan Dong
- Department of Obstetrics and Gynecology, Center for Reproductive Medicine, Qilu Hospital, Shandong University, Ji'nan, Shandong, 250012, P.R. China
| | - Hengfei Li
- School of Computer Science and Technology, Shandong Jianzhu University, Jinan, Shandong, 250101, P.R. China
| | - Yang Yang
- Department of Obstetrics and Gynecology, Center for Reproductive Medicine, Qilu Hospital, Shandong University, Ji'nan, Shandong, 250012, P.R. China
| | - An-Liang Guo
- Department of Obstetrics and Gynecology, Center for Reproductive Medicine, Qilu Hospital, Shandong University, Ji'nan, Shandong, 250012, P.R. China
| | - Lan Chao
- Department of Obstetrics and Gynecology, Center for Reproductive Medicine, Qilu Hospital, Shandong University, Ji'nan, Shandong, 250012, P.R. China.
| |
Collapse
|
18
|
Wang Y, Xiang M, Zhou Y, Zheng N, Zhang J, Zha X, Duan Z, Wang F, Zhang Y, Wang Z, Cao Y, Zhu F. Novel and recurrent hemizygous variants in BCORL1 cause oligoasthenoteratozoospermia by interfering transcription. Andrology 2024. [PMID: 39189935 DOI: 10.1111/andr.13743] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Revised: 07/09/2024] [Accepted: 08/10/2024] [Indexed: 08/28/2024]
Abstract
BACKGROUND Oligoasthenoteratozoospermia (OAT) is a common cause of male infertility, of which the causes remain largely unknown. Recently, BCORL1 was identified as a contributor to male infertility from non-obstructive azoospermia (NOA) to OAT. OBJECTIVES To identify novel and hotspot variants in BCORL1 from infertile men with OAT and reveal their outcomes of assisted reproductive treatments (ARTs). MATERIALS AND METHODS Forty-six infertile men characterized by OAT were recruited from 2017 to 2022. Variants in OAT patients were identified by whole-exome sequencing (WES) and verified by Sanger sequencing. Papanicolaou staining was used for sperm morphology analysis. Pathogenicity of BCORL1 variants were analyzed by bioinformatics analysis, and further confirmed in vitro by using recombinant plasmids and cells. Meanwhile, ARTs were performed on these patients to investigate the appropriate clinical treatment strategy. RESULTS We identified a novel hemizygous missense variant (NM_021946: c.G4171A; p.G1391R) and a recurrent variant (NM_021946: c.T2615G; p.V872G) in BCORL1 from four OAT patients. Notably, routine semen assessment and Papanicolaou staining revealed a special OAT phenotype of patients with BCORL1 variants, whose rare mature sperm characterized by acephalic and abnormal acrosome. Pathogenicity analysis showed the interaction between BCORL1 with histone deacetylases (HDACs) were disrupted after variance, accompanied with epigenetic alterations and finally the orderly transcriptions of spermatogenetic genes were interfering. Besides, clinical record presented the poor outcomes of ARTs in these patients with BCORL1 variants. DISCUSSION AND CONCLUSIONS Our findings further expand the variant spectrum of BCORL1 related to OAT, and provide new evidences that BCORL1 acts as an important transcriptional regulator, participating in epigenetic regulation and directing the expression of key genes throughout spermatogenesis. The outcomes of ARTs will facilitate the genetic counseling and clinical treatment of infertile men with BCORL1 variants in the future.
Collapse
Affiliation(s)
- Yu Wang
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, the Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, the First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
- Department of clinical laboratory, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
- NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract, Anhui Medical University, Hefei, Anhui, China
- Key Laboratory of Population Health Across Life Cycle, Anhui Medical University, Ministry of Education of the People's Republic of China, Hefei, Anhui, China
- Anhui Province Key Laboratory of Reproductive Health and Genetics, Hefei, Anhui, China
| | - Mingfei Xiang
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, the First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
- Department of clinical laboratory, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
- NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract, Anhui Medical University, Hefei, Anhui, China
- Key Laboratory of Population Health Across Life Cycle, Anhui Medical University, Ministry of Education of the People's Republic of China, Hefei, Anhui, China
- Anhui Province Key Laboratory of Reproductive Health and Genetics, Hefei, Anhui, China
- Biopreservation and Artificial Organs, Anhui Provincial Engineering Research Center, Anhui Medical University, Hefei, Anhui, China
| | - Yiru Zhou
- Anhui Provincial Institute of Translational Medicine, Hefei, Anhui, China
| | - Na Zheng
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, the First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
- Department of clinical laboratory, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
- NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract, Anhui Medical University, Hefei, Anhui, China
- Key Laboratory of Population Health Across Life Cycle, Anhui Medical University, Ministry of Education of the People's Republic of China, Hefei, Anhui, China
- Anhui Province Key Laboratory of Reproductive Health and Genetics, Hefei, Anhui, China
- Biopreservation and Artificial Organs, Anhui Provincial Engineering Research Center, Anhui Medical University, Hefei, Anhui, China
| | - Jingjing Zhang
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, the First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
- Department of clinical laboratory, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
- NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract, Anhui Medical University, Hefei, Anhui, China
- Key Laboratory of Population Health Across Life Cycle, Anhui Medical University, Ministry of Education of the People's Republic of China, Hefei, Anhui, China
- Anhui Province Key Laboratory of Reproductive Health and Genetics, Hefei, Anhui, China
- Biopreservation and Artificial Organs, Anhui Provincial Engineering Research Center, Anhui Medical University, Hefei, Anhui, China
| | - Xiaomin Zha
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, the First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
- Department of clinical laboratory, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
- NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract, Anhui Medical University, Hefei, Anhui, China
- Key Laboratory of Population Health Across Life Cycle, Anhui Medical University, Ministry of Education of the People's Republic of China, Hefei, Anhui, China
- Anhui Province Key Laboratory of Reproductive Health and Genetics, Hefei, Anhui, China
- Biopreservation and Artificial Organs, Anhui Provincial Engineering Research Center, Anhui Medical University, Hefei, Anhui, China
| | - Zongliu Duan
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, the First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
- Department of clinical laboratory, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
- NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract, Anhui Medical University, Hefei, Anhui, China
- Key Laboratory of Population Health Across Life Cycle, Anhui Medical University, Ministry of Education of the People's Republic of China, Hefei, Anhui, China
- Anhui Province Key Laboratory of Reproductive Health and Genetics, Hefei, Anhui, China
- Biopreservation and Artificial Organs, Anhui Provincial Engineering Research Center, Anhui Medical University, Hefei, Anhui, China
| | - Fengsong Wang
- School of Life Science, Anhui Medical University, Hefei, Anhui, China
| | - Ying Zhang
- Department of Obstetrics and Gynecology, the First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Zhongxin Wang
- Anhui Provincial Institute of Translational Medicine, Hefei, Anhui, China
| | - Yunxia Cao
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, the First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
- Department of clinical laboratory, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
- NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract, Anhui Medical University, Hefei, Anhui, China
- Key Laboratory of Population Health Across Life Cycle, Anhui Medical University, Ministry of Education of the People's Republic of China, Hefei, Anhui, China
- Anhui Province Key Laboratory of Reproductive Health and Genetics, Hefei, Anhui, China
- Biopreservation and Artificial Organs, Anhui Provincial Engineering Research Center, Anhui Medical University, Hefei, Anhui, China
| | - Fuxi Zhu
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, the Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, the First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
- Department of clinical laboratory, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
- NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract, Anhui Medical University, Hefei, Anhui, China
- Key Laboratory of Population Health Across Life Cycle, Anhui Medical University, Ministry of Education of the People's Republic of China, Hefei, Anhui, China
- Anhui Province Key Laboratory of Reproductive Health and Genetics, Hefei, Anhui, China
| |
Collapse
|
19
|
Wan X, Hu H, Sun J, Meng F, Gong F, Lin G, Liao H, Zheng W. Identification of novel compound heterozygous ZFP36L2 variants implicated in oocyte maturation defects and female infertility. J Assist Reprod Genet 2024; 41:1955-1963. [PMID: 38829516 PMCID: PMC11339011 DOI: 10.1007/s10815-024-03154-1] [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: 02/08/2024] [Accepted: 05/22/2024] [Indexed: 06/05/2024] Open
Abstract
PURPOSE To explore the pathogenesis of oocyte maturation defects. METHODS Whole exome sequencing was conducted to identify potential variants, which were then confirmed within the pedigree through Sanger sequencing. The functional characterization of the identified variants responsible for the disease, including their subcellular localization, protein levels, and interactions with other proteins, was verified through transient transfection in HeLa cells in vitro. Additionally, we employed real-time RT-PCR and single-cell RNA sequencing to examine the impact of ZFP36L2 pathogenic variants on mRNA metabolism in both HeLa cells and mouse or human oocytes. RESULTS A novel compound heterozygous variant in ZFP36L2 (c.186T > G, p.His62Gln and c.869 C > T, p.Pro290Leu) was discovered in a patient with oocyte maturation defects. Our findings indicate that these variants lead to compromised binding capacity of the ZFP36L2-CONT6L complex and impaired mRNA degradation in HeLa cells and mouse oocytes. Furthermore, we characterized the changes in the human oocyte transcriptome associated with ZFP36L2 variants, with a particular emphasis on cell division, mitochondrial function, and ribosome metabolism. CONCLUSIONS This study broadens the mutation spectrum of ZFP36L2 and constitutes the first report of human oocyte transcriptome alterations linked to ZFP36L2 variants. In conjunction with existing knowledge of ZFP36L2, our research lays the groundwork for genetic counseling aimed at addressing female infertility.
Collapse
Affiliation(s)
- Xian Wan
- The Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, China
- Hengyang Nanhua-Xinghui Reproductive Health Hospital, Hengyang, China
| | - Huiling Hu
- Clinical Research Center for Reproduction and Genetics in Hunan Province, Reproductive and Genetic Hospital of CITIC-XIANGYA, Changsha, 410008, China
| | - Jiaqi Sun
- Center for Medical Genetics, School of Life Sciences, Central South University, Changsha, China
| | - Fei Meng
- Clinical Research Center for Reproduction and Genetics in Hunan Province, Reproductive and Genetic Hospital of CITIC-XIANGYA, Changsha, 410008, China
| | - Fei Gong
- Clinical Research Center for Reproduction and Genetics in Hunan Province, Reproductive and Genetic Hospital of CITIC-XIANGYA, Changsha, 410008, China
| | - Ge Lin
- Clinical Research Center for Reproduction and Genetics in Hunan Province, Reproductive and Genetic Hospital of CITIC-XIANGYA, Changsha, 410008, China
| | - Hongqing Liao
- The Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, China.
- Hengyang Nanhua-Xinghui Reproductive Health Hospital, Hengyang, China.
| | - Wei Zheng
- Center for Medical Genetics, School of Life Sciences, Central South University, Changsha, China.
- Clinical Research Center for Reproduction and Genetics in Hunan Province, Reproductive and Genetic Hospital of CITIC-XIANGYA, Changsha, 410008, China.
| |
Collapse
|
20
|
Hu HY, Zhang GH, Deng WF, Wei TY, Feng ZK, Li CX, Li SJ, Liu JE, Tian YP. Novel PATL2 variants cause female infertility with oocyte maturation defect. J Assist Reprod Genet 2024; 41:1965-1976. [PMID: 38954294 PMCID: PMC11339215 DOI: 10.1007/s10815-024-03150-5] [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: 01/23/2024] [Accepted: 05/22/2024] [Indexed: 07/04/2024] Open
Abstract
PURPOSE Oocyte maturation defect (OOMD) is a rare cause of in vitro fertilization failure characterized by the production of immature oocytes. Compound heterozygous or homozygous PATL2 mutations have been associated with oocyte arrest at the germinal vesicle (GV), metaphase I (MI), and metaphase II (MII) stages, as well as morphological changes. METHODS In this study, we recruited three OOMD cases and conducted a comprehensive multiplatform laboratory investigation. RESULTS Whole exome sequence (WES) revealed four diagnostic variants in PATL2, nonsense mutation c.709C > T (p.R237*) and frameshift mutation c.1486_1487delinsT (p.A496Sfs*4) were novel mutations that have not been reported previously. Furthermore, the pathogenicity of these variants was predicted using in silico analysis, which indicated detrimental effects. Molecular dynamic analysis suggested that the A496S variant disrupted the hydrophobic segment, leading to structural changes that affected the overall protein folding and stability. Additionally, biochemical and molecular experiments were conducted on cells transfected with wild-type (WT) or mutant PATL2 (p.R237* and p.A496Sfs*4) plasmid vectors. CONCLUSIONS The results demonstrated that PATL2A496Sfs*4 and PATL2R237* had impacts on protein size and expression level. Interestingly, expression levels of specific genes involved in oocyte maturation and early embryonic development were found to be simultaneously deregulated. The findings in our study expand the variation spectrum of the PATL2 gene, provide solid evidence for counseling on future pregnancies in affected families, strongly support the application of in the diagnosis of OOMD, and contribute to the understanding of PATL2 function.
Collapse
Affiliation(s)
- Hua-Ying Hu
- Birth Defects Prevention and Control Technology Research Center, Medical Innovation Research Division of Chinese, PLA General Hospital, Beijing, 100853, China
| | - Ge-Han Zhang
- Translational Medicine Research Center, Medical Innovation Research Division of Chinese, PLA General Hospital, Beijing, China
| | - Wei-Fen Deng
- Shenzhen Hengsheng Hospital, Shenzhen, Guangdong, China
| | - Tian-Ying Wei
- Jiaen Genetics Laboratory, Beijing Jiaen Hospital, Beijing, 100191, China
| | - Zhan-Ke Feng
- Jiaen Genetics Laboratory, Beijing Jiaen Hospital, Beijing, 100191, China
| | - Cun-Xi Li
- Jiaen Genetics Laboratory, Beijing Jiaen Hospital, Beijing, 100191, China
| | - Song Jun Li
- The Reproduction Medical Center, The Third Affiliated Hospital of Shenzhen University, Shenzhen, 518001, Guangdong, China.
| | - Jia-En Liu
- Jiaen Genetics Laboratory, Beijing Jiaen Hospital, Beijing, 100191, China.
| | - Ya-Ping Tian
- Birth Defects Prevention and Control Technology Research Center, Medical Innovation Research Division of Chinese, PLA General Hospital, Beijing, 100853, China.
| |
Collapse
|
21
|
Li W, Li H, Zha C, Che B, Yu Y, Yang J, Li T. Lipids, lipid-modified drug target genes, and the risk of male infertility: a Mendelian randomization study. Front Endocrinol (Lausanne) 2024; 15:1392533. [PMID: 39114294 PMCID: PMC11303150 DOI: 10.3389/fendo.2024.1392533] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Accepted: 06/28/2024] [Indexed: 08/10/2024] Open
Abstract
Background Previous observational studies have reported a possible association between circulating lipids and lipid-lowering drugs and male infertility (MIF), as well as the mediating role of circulating vitamin D. Then, due to issues such as bias, reverse causality, and residual confounding, inferring causal relationships from these studies may be challenging. Therefore, this study aims to explore the effects of circulating lipids and lipid-lowering drugs on MIF through Mendelian randomization (MR) analysis and evaluate the mediating role of vitamin D. Method Genetic variations related to lipid traits and the lipid-lowering effect of lipid modification targets are extracted from the Global Alliance for Lipid Genetics Genome-Wide Association Study. The summary statistics for MIF are from the FinnGen 9th edition. Using quantitative expression feature loci data from relevant organizations to obtain genetic variations related to gene expression level, further to explore the relationship between these target gene expression levels and MIF risk. Two-step MR analysis is used to explore the mediating role of vitamin D. Multiple sensitivity analysis methods (co-localization analysis, Egger intercept test, Cochrane's Q test, pleiotropy residuals and outliers (MR-PRESSO), and the leave-one-out method) are used to demonstrate the reliability of our results. Result In our study, we observed that lipid modification of four lipid-lowering drug targets was associated with MIF risk, the LDLR activator (equivalent to a 1-SD decrease in LDL-C) (OR=1.94, 95% CI 1.14-3.28, FDR=0.040), LPL activator (equivalent to a 1-SD decrease in TG) (OR=1.86, 95% CI 1.25-2.76, FDR=0.022), and CETP inhibitor (equivalent to a 1-SD increase in HDL-C) (OR=1.28, 95% CI 1.07-1.53, FDR=0.035) were associated with a higher risk of MIF. The HMGCR inhibitor (equivalent to a 1-SD decrease in LDL-C) was associated with a lower risk of MIF (OR=0.38, 95% CI 0.17-0.83, FDR=0.39). Lipid-modifying effects of three targets were partially mediated by serum vitamin D levels. Mediation was 0.035 (LDLR activator), 0.012 (LPL activator), and 0.030 (CETP inhibitor), with mediation ratios of 5.34% (LDLR activator), 1.94% (LPL activator), and 12.2% (CETP inhibitor), respectively. In addition, there was no evidence that lipid properties and lipid modification effects of six other lipid-lowering drug targets were associated with MIF risk. Multiple sensitivity analysis methods revealed insignificant evidence of bias arising from pleiotropy or genetic confounding. Conclusion This study did not support lipid traits (LDL-C, HDL-C, TG, Apo-A1, and Apo-B) as pathogenic risk factors for MIF. It emphasized that LPL, LDLR, CETP, and HMGCR were promising drug targets for improving male fertility.
Collapse
Affiliation(s)
- Wei Li
- Department of Urology, the Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Hu Li
- Emergency Department, Affiliated Hospital of Binzhou Medical College, Binzhou, China
| | - Cheng Zha
- Department of Urology, the Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Bangwei Che
- Department of Urology, The First Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, China
| | - Ying Yu
- Department of Urology, the Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Jianjun Yang
- Department of Urology, the Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Tao Li
- Department of Urology, the Affiliated Hospital of Guizhou Medical University, Guiyang, China
| |
Collapse
|
22
|
Guo L, Yin S, Wei H, Peng J. No evidence of genetic causation between iron and infertility: a Mendelian randomization study. Front Nutr 2024; 11:1390618. [PMID: 39104757 PMCID: PMC11298439 DOI: 10.3389/fnut.2024.1390618] [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: 02/23/2024] [Accepted: 07/08/2024] [Indexed: 08/07/2024] Open
Abstract
Background Observational studies have explored the impact of iron homeostasis on infertility; however, establishing definitive causal relationships remains challenging. This study utilized a two-sample Mendelian randomization approach to investigate the potential causal relationship between iron status and infertility. Materials and methods Four indicators of iron status-serum iron, ferritin, transferrin saturation, and total iron binding capacity, were considered as exposure factors. Infertility was the outcome variable for both men and women. Robust causality was assessed using the primary inverse-variance-weighted method, complemented by three supplementary Mendelian randomization approaches. Sensitivity analyses were performed to enhance the precision and reliability of the results. Results No statistically significant associations were identified between the four indicators of iron status and infertility. These results remained consistent across multiple Mendelian randomization methodologies. Conclusion In conclusion, there is no evidence of a genetic causal relationship between iron status and infertility. Nevertheless, this does not preclude the possibility of a connection between iron status and infertility at different mechanistic levels.
Collapse
Affiliation(s)
- Liangliang Guo
- Department of Animal Nutrition and Feed Science, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, China
| | - Shengnan Yin
- Department of Animal Nutrition and Feed Science, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, China
| | - Hongkui Wei
- Department of Animal Nutrition and Feed Science, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, China
| | - Jian Peng
- Department of Animal Nutrition and Feed Science, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, China
- The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, China
- Frontiers Science Center for Animal Breeding and Sustainable Production, Wuhan, China
| |
Collapse
|
23
|
Li W, Huang X, Wei Y, Yin T, Diao L. Connecting the dots: the role of fatigue in female infertility. Reprod Biol Endocrinol 2024; 22:66. [PMID: 38849828 PMCID: PMC11157719 DOI: 10.1186/s12958-024-01235-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Accepted: 05/21/2024] [Indexed: 06/09/2024] Open
Abstract
Fatigue, an increasingly acknowledged symptom in various chronic diseases, has garnered heightened attention, during the medical era of bio-psycho-social model. Its persistence not only significantly compromises an individual's quality of life but also correlates with chronic organ damage. Surprisingly, the intricate relationship between fatigue and female reproductive health, specifically infertility, remains largely unexplored. Our exploration into the existing body of evidence establishes a compelling link between fatigue with uterine and ovarian diseases, as well as conditions associated with infertility, such as rheumatism. This observation suggests a potentially pivotal role of fatigue in influencing overall female fertility. Furthermore, we propose a hypothetical mechanism elucidating the impact of fatigue on infertility from multiple perspectives, postulating that neuroendocrine, neurotransmitter, inflammatory immune, and mitochondrial dysfunction resulting from fatigue and its co-factors may further contribute to endocrine disorders, menstrual irregularities, and sexual dysfunction, ultimately leading to infertility. In addition to providing this comprehensive theoretical framework, we summarize anti-fatigue strategies and accentuate current knowledge gaps. By doing so, our aim is to offer novel insights, stimulate further research, and advance our understanding of the crucial interplay between fatigue and female reproductive health.
Collapse
Grants
- 82371684, 82271672 General Program of the National Natural Science Foundation of China
- 82371684, 82271672 General Program of the National Natural Science Foundation of China
- 82371684, 82271672 General Program of the National Natural Science Foundation of China
- 82371684, 82271672 General Program of the National Natural Science Foundation of China
- 82371684, 82271672 General Program of the National Natural Science Foundation of China
- JCRCWL-2022-001 the Interdisciplinary Innovative Talents Foundation from Renmin Hospital of Wuhan University
- JCRCWL-2022-001 the Interdisciplinary Innovative Talents Foundation from Renmin Hospital of Wuhan University
- JCRCWL-2022-001 the Interdisciplinary Innovative Talents Foundation from Renmin Hospital of Wuhan University
- JCRCWL-2022-001 the Interdisciplinary Innovative Talents Foundation from Renmin Hospital of Wuhan University
- JCRCWL-2022-001 the Interdisciplinary Innovative Talents Foundation from Renmin Hospital of Wuhan University
- 2022A1515010650, 2023A1515011675 the General Program of the Natural Science Foundation of Guangdong Province
- 2022A1515010650, 2023A1515011675 the General Program of the Natural Science Foundation of Guangdong Province
- 2022A1515010650, 2023A1515011675 the General Program of the Natural Science Foundation of Guangdong Province
- 2022A1515010650, 2023A1515011675 the General Program of the Natural Science Foundation of Guangdong Province
- 2022A1515010650, 2023A1515011675 the General Program of the Natural Science Foundation of Guangdong Province
Collapse
Affiliation(s)
- Wenzhu Li
- Reproductive Medical Center, Renmin Hospital of Wuhan University and Hubei Clinic Research Center for Assisted Reproductive Technology and Embryonic Development, Wuhan, 430060, China
| | - Xiaoyan Huang
- Department of Rheumatology, The University of Hong Kong- Shenzhen Hospital, Shenzhen, 518053, China
| | - Yiqiu Wei
- Reproductive Medical Center, Renmin Hospital of Wuhan University and Hubei Clinic Research Center for Assisted Reproductive Technology and Embryonic Development, Wuhan, 430060, China
| | - Tailang Yin
- Reproductive Medical Center, Renmin Hospital of Wuhan University and Hubei Clinic Research Center for Assisted Reproductive Technology and Embryonic Development, Wuhan, 430060, China.
| | - Lianghui Diao
- Shenzhen Key Laboratory of Reproductive Immunology for Peri-implantation, Shenzhen Zhongshan Institute for Reproductive Medicine and Genetics, Shenzhen Zhongshan Obstetrics & Gynecology Hospital (formerly Shenzhen Zhongshan Urology Hospital), Shenzhen, 518045, China.
- Guangdong Engineering Technology Research Center of Reproductive Immunology for Peri- implantation, Shenzhen, 518045, China.
| |
Collapse
|
24
|
Zhang C, Zhang Y, Chang Z, Li C. Sperm YOLOv8E-TrackEVD: A Novel Approach for Sperm Detection and Tracking. SENSORS (BASEL, SWITZERLAND) 2024; 24:3493. [PMID: 38894284 PMCID: PMC11175353 DOI: 10.3390/s24113493] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Revised: 05/21/2024] [Accepted: 05/24/2024] [Indexed: 06/21/2024]
Abstract
Male infertility is a global health issue, with 40-50% attributed to sperm abnormalities. The subjectivity and irreproducibility of existing detection methods pose challenges to sperm assessment, making the design of automated semen analysis algorithms crucial for enhancing the reliability of sperm evaluations. This paper proposes a comprehensive sperm tracking algorithm (Sperm YOLOv8E-TrackEVD) that combines an enhanced YOLOv8 small object detection algorithm (SpermYOLOv8-E) with an improved DeepOCSORT tracking algorithm (SpermTrack-EVD) to detect human sperm in a microscopic field of view and track healthy sperm in a sample in a short period effectively. Firstly, we trained the improved YOLOv8 model on the VISEM-Tracking dataset for accurate sperm detection. To enhance the detection of small sperm objects, we introduced an attention mechanism, added a small object detection layer, and integrated the SPDConv and Detect_DyHead modules. Furthermore, we used a new distance metric method and chose IoU loss calculation. Ultimately, we achieved a 1.3% increase in precision, a 1.4% increase in recall rate, and a 2.0% improvement in mAP@0.5:0.95. We applied SpermYOLOv8-E combined with SpermTrack-EVD for sperm tracking. On the VISEM-Tracking dataset, we achieved 74.303% HOTA and 71.167% MOTA. These results show the effectiveness of the designed Sperm YOLOv8E-TrackEVD approach in sperm tracking scenarios.
Collapse
Affiliation(s)
| | | | - Zhanyuan Chang
- College of Information, Mechanical and Electrical Engineering, Shanghai Normal University, Shanghai 200234, China; (C.Z.); (Y.Z.); (C.L.)
| | | |
Collapse
|
25
|
Pascuali N, Pu Y, Waye AA, Pearl S, Martin D, Sutton A, Shikanov A, Veiga-Lopez A. Evaluation of Lipids and Lipid-Related Transcripts in Human and Ovine Theca Cells and an in Vitro Mouse Model Exposed to the Obesogen Chemical Tributyltin. ENVIRONMENTAL HEALTH PERSPECTIVES 2024; 132:47009. [PMID: 38630605 PMCID: PMC11023052 DOI: 10.1289/ehp13955] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Revised: 02/22/2024] [Accepted: 03/18/2024] [Indexed: 04/19/2024]
Abstract
BACKGROUND Exposure to obesogenic chemicals has been reported to result in enhanced adipogenesis, higher adipose tissue accumulation, and reduced ovarian hormonal synthesis and follicular function. We have reported that organotins [tributyltin (TBT) and triphenyltin (TPT)] dysregulate cholesterol trafficking in ovarian theca cells, but, whether organotins also exert lipogenic effects on ovarian cells remains unexplored. OBJECTIVE We investigated if environmentally relevant exposures to organotins [TBT, TPT, or dibutyltin (DBT)] induce lipid dysregulation in ovarian theca cells and the role of the liver X receptor (LXR) in this effect. We also tested the effect of TBT on oocyte maturation and neutral lipid accumulation, and lipid-related transcript expression in cumulus cells and preimplantation embryos. METHODS Primary theca cell cultures derived from human and ovine ovaries were exposed to TBT, TPT, or DBT (1, 10, or 50 ng / ml ). The effect of these chemical exposures on neutral lipid accumulation, lipid abundance and composition, lipid homeostasis-related gene expression, and cytokine secretion was evaluated using liquid chromatography-mass spectrometry (LC-MS), inhibitor-based methods, cytokine secretion, and lipid ontology analyses. We also exposed murine cumulus-oocyte complexes to TBT and evaluated oocyte maturation, embryo development, and lipid homeostasis-related mRNA expression in cumulus cells and blastocysts. RESULTS Exposure to TBT resulted in higher intracellular neutral lipids in human and ovine primary theca cells. In ovine theca cells, this effect was dose-dependent, independent of cell stage, and partially mediated by LXR. DBT and TPT resulted in higher intracellular neutral lipids but to a lesser extent in comparison with TBT. More than 140 lipids and 9 cytokines were dysregulated in TBT-exposed human theca cells. Expression of genes involved in lipogenesis and fatty acid synthesis were higher in theca cells, as well as in cumulus cells and blastocysts exposed to TBT. However, TBT did not impact the rates of oocyte maturation or blastocyst development. DISCUSSION TBT induced dyslipidemia in primary human and ovine theca cells, which may be responsible for some of the TBT-induced fertility dysregulations reported in rodent models of TBT exposure. https://doi.org/10.1289/EHP13955.
Collapse
Affiliation(s)
- Natalia Pascuali
- Department of Pathology, University of Illinois Chicago, Chicago, Illinois, USA
| | - Yong Pu
- Department of Pathology, University of Illinois Chicago, Chicago, Illinois, USA
| | - Anita A. Waye
- Department of Pathology, University of Illinois Chicago, Chicago, Illinois, USA
| | - Sarah Pearl
- Department of Obstetrics and Gynecology, Sparrow Health System, Lansing, Michigan, USA
| | - Denny Martin
- Department of Obstetrics and Gynecology, Sparrow Health System, Lansing, Michigan, USA
| | - Allison Sutton
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, Michigan, USA
| | - Ariella Shikanov
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, Michigan, USA
| | - Almudena Veiga-Lopez
- Department of Pathology, University of Illinois Chicago, Chicago, Illinois, USA
- The Chicago Center for Health and Environment, University of Illinois Chicago, Chicago, Illinois, USA
| |
Collapse
|
26
|
Wei SM, Gregory MD, Nash T, de Abreu e Gouvêa A, Mervis CB, Cole KM, Garvey MH, Kippenhan JS, Eisenberg DP, Kolachana B, Schmidt PJ, Berman KF. Altered pubertal timing in 7q11.23 copy number variations and associated genetic mechanisms. iScience 2024; 27:109113. [PMID: 38375233 PMCID: PMC10875153 DOI: 10.1016/j.isci.2024.109113] [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: 09/05/2023] [Revised: 11/20/2023] [Accepted: 01/31/2024] [Indexed: 02/21/2024] Open
Abstract
Pubertal timing, including age at menarche (AAM), is a heritable trait linked to lifetime health outcomes. Here, we investigate genetic mechanisms underlying AAM by combining genome-wide association study (GWAS) data with investigations of two rare genetic conditions clinically associated with altered AAM: Williams syndrome (WS), a 7q11.23 hemideletion characterized by early puberty; and duplication of the same genes (7q11.23 Duplication syndrome [Dup7]) characterized by delayed puberty. First, we confirm that AAM-derived polygenic scores in typically developing children (TD) explain a modest amount of variance in AAM (R2 = 0.09; p = 0.04). Next, we demonstrate that 7q11.23 copy number impacts AAM (WS < TD < Dup7; p = 1.2x10-8, η2 = 0.45) and pituitary volume (WS < TD < Dup7; p = 3x10-5, ηp2 = 0.2) with greater effect sizes. Finally, we relate an AAM-GWAS signal in 7q11.23 to altered expression in postmortem brains of STAG3L2 (p = 1.7x10-17), a gene we also find differentially expressed with 7q11.23 copy number (p = 0.03). Collectively, these data explicate the role of 7q11.23 in pubertal onset, with STAG3L2 and pituitary development as potential mediators.
Collapse
Affiliation(s)
- Shau-Ming Wei
- Behavioral Endocrinology Branch, National Institute of Mental Health, Intramural Research Program, National Institutes of Health, Bethesda, MD, USA
- Section on Integrative Neuroimaging, Clinical and Translational Neuroscience Branch, National Institute of Mental Health, Intramural Research Program, National Institutes of Health, Bethesda, MD, USA
| | - Michael D. Gregory
- Section on Integrative Neuroimaging, Clinical and Translational Neuroscience Branch, National Institute of Mental Health, Intramural Research Program, National Institutes of Health, Bethesda, MD, USA
| | - Tiffany Nash
- Section on Integrative Neuroimaging, Clinical and Translational Neuroscience Branch, National Institute of Mental Health, Intramural Research Program, National Institutes of Health, Bethesda, MD, USA
| | - Andrea de Abreu e Gouvêa
- Section on Integrative Neuroimaging, Clinical and Translational Neuroscience Branch, National Institute of Mental Health, Intramural Research Program, National Institutes of Health, Bethesda, MD, USA
| | - Carolyn B. Mervis
- Neurodevelopmental Sciences Laboratory, Department of Psychological and Brain Sciences, University of Louisville, Louisville, KY, USA
| | - Katherine M. Cole
- Behavioral Endocrinology Branch, National Institute of Mental Health, Intramural Research Program, National Institutes of Health, Bethesda, MD, USA
- Section on Integrative Neuroimaging, Clinical and Translational Neuroscience Branch, National Institute of Mental Health, Intramural Research Program, National Institutes of Health, Bethesda, MD, USA
| | - Madeline H. Garvey
- Section on Integrative Neuroimaging, Clinical and Translational Neuroscience Branch, National Institute of Mental Health, Intramural Research Program, National Institutes of Health, Bethesda, MD, USA
| | - J. Shane Kippenhan
- Section on Integrative Neuroimaging, Clinical and Translational Neuroscience Branch, National Institute of Mental Health, Intramural Research Program, National Institutes of Health, Bethesda, MD, USA
| | - Daniel P. Eisenberg
- Section on Integrative Neuroimaging, Clinical and Translational Neuroscience Branch, National Institute of Mental Health, Intramural Research Program, National Institutes of Health, Bethesda, MD, USA
| | - Bhaskar Kolachana
- Human Brain Collection Core, National Institute of Mental Health, Intramural Research Program, National Institutes of Health, Bethesda, MD, USA
| | - Peter J. Schmidt
- Behavioral Endocrinology Branch, National Institute of Mental Health, Intramural Research Program, National Institutes of Health, Bethesda, MD, USA
| | - Karen F. Berman
- Section on Integrative Neuroimaging, Clinical and Translational Neuroscience Branch, National Institute of Mental Health, Intramural Research Program, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
27
|
Li Q, Hu Z, He J, Liu X, Liu Y, Wei J, Wu B, Lu X, He H, Zhang Y, He J, Li M, Wu C, Lv L, Wang Y, Zhou L, Zhang Q, Zhang J, Cheng X, Shao H, Lu X. Deciphering the comprehensive knowledgebase landscape featuring infertility with IDDB Xtra. Comput Biol Med 2024; 170:108105. [PMID: 38330823 DOI: 10.1016/j.compbiomed.2024.108105] [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: 12/20/2023] [Revised: 01/15/2024] [Accepted: 02/04/2024] [Indexed: 02/10/2024]
Abstract
Infertility affects ∼15% of couples globally and half of cases are related to genetic disorders. Despite growing data and unprecedented improvements in high-throughput sequencing technologies, accumulated fertility-related issues concerning genetic diagnosis and potential treatment are urgent to be solved. However, there is a lack of comprehensive platforms that characterise various infertility-related records to provide research applications for exploring infertility in-depth and genetic counselling of infertility couple. To solve this problem, we provide IDDB Xtra by further integrating phenotypic manifestations, genomic datasets, epigenetics, modulators in collaboration with numerous interactive tools into our previous infertility database, IDDB. IDDB Xtra houses manually-curated 2369 genes of human and nine model organisms, 273 chromosomal abnormalities, 884 phenotypes, 60 genomic datasets, 464 epigenetic records, 1144 modulators relevant to infertility diagnosis and treatment. Additionally, IDDB Xtra incorporated customized graphical applications for researchers and clinicians to decipher in-depth disease mechanisms from the perspectives of developmental atlas, mutation effects, and clinical manifestations. Users can browse genes across developmental stages of human and mouse, filter candidate genes, mine potential variants and retrieve infertility biomedical network in an intuitive web interface. In summary, IDDB Xtra not only captures valuable research and data, but also provides useful applications to facilitate the genetic counselling and drug discovery of infertility. IDDB Xtra is freely available at https://mdl.shsmu.edu.cn/IDDB/and http://www.allostery.net/IDDB.
Collapse
Affiliation(s)
- Qian Li
- Department of Assisted Reproduction, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine (SJTU-SM), Shanghai, 200011, China; Medicinal Bioinformatics Center, Shanghai Jiao Tong University School of Medicine (SJTU-SM), Shanghai, 200025, China
| | - Zhijie Hu
- Department of Assisted Reproduction, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine (SJTU-SM), Shanghai, 200011, China
| | - Jiayin He
- Department of Assisted Reproduction, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine (SJTU-SM), Shanghai, 200011, China; Medicinal Bioinformatics Center, Shanghai Jiao Tong University School of Medicine (SJTU-SM), Shanghai, 200025, China
| | - Xinyi Liu
- Medicinal Bioinformatics Center, Shanghai Jiao Tong University School of Medicine (SJTU-SM), Shanghai, 200025, China
| | - Yini Liu
- Department of Assisted Reproduction, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine (SJTU-SM), Shanghai, 200011, China; Medicinal Bioinformatics Center, Shanghai Jiao Tong University School of Medicine (SJTU-SM), Shanghai, 200025, China
| | - Jiale Wei
- Medicinal Bioinformatics Center, Shanghai Jiao Tong University School of Medicine (SJTU-SM), Shanghai, 200025, China
| | - Binjian Wu
- Medicinal Bioinformatics Center, Shanghai Jiao Tong University School of Medicine (SJTU-SM), Shanghai, 200025, China
| | - Xun Lu
- Medicinal Bioinformatics Center, Shanghai Jiao Tong University School of Medicine (SJTU-SM), Shanghai, 200025, China
| | - Hongxi He
- Department of Assisted Reproduction, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine (SJTU-SM), Shanghai, 200011, China; School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510000, China
| | - Yuqi Zhang
- Department of Assisted Reproduction, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine (SJTU-SM), Shanghai, 200011, China
| | - Jixiao He
- Medicinal Bioinformatics Center, Shanghai Jiao Tong University School of Medicine (SJTU-SM), Shanghai, 200025, China
| | - Mingyu Li
- Medicinal Bioinformatics Center, Shanghai Jiao Tong University School of Medicine (SJTU-SM), Shanghai, 200025, China
| | - Chengwei Wu
- Medicinal Bioinformatics Center, Shanghai Jiao Tong University School of Medicine (SJTU-SM), Shanghai, 200025, China
| | - Lijun Lv
- Medicinal Bioinformatics Center, Shanghai Jiao Tong University School of Medicine (SJTU-SM), Shanghai, 200025, China
| | - Yang Wang
- Medicinal Bioinformatics Center, Shanghai Jiao Tong University School of Medicine (SJTU-SM), Shanghai, 200025, China
| | - Linxuan Zhou
- Medicinal Bioinformatics Center, Shanghai Jiao Tong University School of Medicine (SJTU-SM), Shanghai, 200025, China
| | - Quan Zhang
- Medicinal Bioinformatics Center, Shanghai Jiao Tong University School of Medicine (SJTU-SM), Shanghai, 200025, China
| | - Jian Zhang
- Medicinal Bioinformatics Center, Shanghai Jiao Tong University School of Medicine (SJTU-SM), Shanghai, 200025, China; School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510000, China.
| | - Xiaoyue Cheng
- Center for Reproductive Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200135, China.
| | - Hongfang Shao
- Center of Reproductive Medicine, Department of Gynecology and Obstetrics, Shanghai Jiao Tong University School of Medicine Affiliated Sixth People's Hospital, Shanghai, 200233, China.
| | - Xuefeng Lu
- Department of Assisted Reproduction, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine (SJTU-SM), Shanghai, 200011, China.
| |
Collapse
|
28
|
Ge C, Zhang B, Mao Y, Hong Z, Zhou C, Wang Y, Wang M, Ma L. Effects of atosiban on clinical outcome in frozen-thawed embryo transfer: a propensity score matching study. Arch Gynecol Obstet 2024; 309:1101-1106. [PMID: 38240770 DOI: 10.1007/s00404-023-07289-x] [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: 09/16/2023] [Accepted: 11/06/2023] [Indexed: 02/16/2024]
Abstract
PURPOSE To evaluate the effects of atosiban on clinical outcomes in patients undergoing frozen-thawed embryo transfer. METHODS The clinical data of 1093 infertile patients who underwent frozen-thawed embryo transfer in our center from January 2019 to December 2020 were retrospectively analyzed (control, 418; atosiban, 675). Propensity score matching (PSM) analysis identified 400 matched pairs of patients. The implantation rate, clinical pregnancy rate, live birth rate, biochemical pregnancy rate, abortion rate, multiple pregnancy rate, and ectopic pregnancy rate between the two groups were compared. RESULTS Before PSM, patients differed by infertility factors, number of transferred embryos, and endometrial preparation protocol (P < 0.05). After PSM, characteristics were similar in corresponding patients of the atosiban and control groups. After propensity score matching, we found that there was no significant difference in the implantation rate, clinical pregnancy rate, live birth rate, biochemical pregnancy rate, abortion rate, multiple pregnancy rate, and ectopic pregnancy rate in atosiban and control group (P > 0.05). CONCLUSION Atosiban did not improve the clinical outcomes of infertile patients with frozen-thawed embryo transfer.
Collapse
Affiliation(s)
- Caiyun Ge
- Reproductive Medicine Center, Zhongnan Hospital of Wuhan University, Hubei Province, No. 169, Wuchang District, East Lake Road, Wuhan City, 430071, China
| | - Bo Zhang
- Reproductive Medicine Center, Zhongnan Hospital of Wuhan University, Hubei Province, No. 169, Wuchang District, East Lake Road, Wuhan City, 430071, China
| | - Yanhong Mao
- Reproductive Medicine Center, Zhongnan Hospital of Wuhan University, Hubei Province, No. 169, Wuchang District, East Lake Road, Wuhan City, 430071, China
| | - Zhidan Hong
- Reproductive Medicine Center, Zhongnan Hospital of Wuhan University, Hubei Province, No. 169, Wuchang District, East Lake Road, Wuhan City, 430071, China
| | - Chun Zhou
- Reproductive Medicine Center, Zhongnan Hospital of Wuhan University, Hubei Province, No. 169, Wuchang District, East Lake Road, Wuhan City, 430071, China
| | - Yan Wang
- Reproductive Medicine Center, Zhongnan Hospital of Wuhan University, Hubei Province, No. 169, Wuchang District, East Lake Road, Wuhan City, 430071, China
| | - Mei Wang
- Reproductive Medicine Center, Zhongnan Hospital of Wuhan University, Hubei Province, No. 169, Wuchang District, East Lake Road, Wuhan City, 430071, China
| | - Ling Ma
- Reproductive Medicine Center, Zhongnan Hospital of Wuhan University, Hubei Province, No. 169, Wuchang District, East Lake Road, Wuhan City, 430071, China.
| |
Collapse
|
29
|
Yalcin Z, Liang M, Abdelrazek IM, Friedrich C, Bareke E, Nabil A, Tüttelmann F, Majewski J, Abdalla E, Tan SL, Slim R. A report of two homozygous TERB1 protein-truncating variants in two unrelated women with primary infertility. J Assist Reprod Genet 2024; 41:751-756. [PMID: 38277113 PMCID: PMC10957843 DOI: 10.1007/s10815-024-03031-x] [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: 09/25/2023] [Accepted: 01/12/2024] [Indexed: 01/27/2024] Open
Abstract
PURPOSE To investigate the genetic etiology of patients with female infertility. METHODS Whole Exome Sequencing was performed on genomic DNA extracted from the patient's blood. Exome data were filtered for damaging rare biallelic variants in genes with possible roles in reproduction. Sanger sequencing was used to validate the selected variants and segregate them in family members. RESULTS A novel homozygous likely pathogenic variant, c.626G>A, p.Trp209*, was identified in the TERB1 gene of the patient. Additionally, we report a second homozygous pathogenic TERB1 variant, c.1703C>G, p.Ser568*, in an infertile woman whose azoospermic brother was previously described to be homozygous for her variant. CONCLUSIONS Here, we report for the first time two homozygous likely pathogenic and pathogenic TERB1 variants, c.626G>A, p.Trp209* and c.1703C>G, p.Ser568*, respectively, in two unrelated women with primary infertility. TERB1 is known to play an essential role in homologous chromosome movement, synapsis, and recombination during the meiotic prophase I and has an established role in male infertility in humans. Our data add TERB1 to the shortlist of Meiosis I genes associated with human infertility in both sexes.
Collapse
Affiliation(s)
- Zeynep Yalcin
- Department of Human Genetics, McGill University Health Centre, Montreal, QC, Canada
| | - Manqi Liang
- Department of Human Genetics, McGill University Health Centre, Montreal, QC, Canada
| | - Ibrahim M Abdelrazek
- Department of Human Genetics, Medical Research Institute, Alexandria University, Alexandria, Egypt
| | - Corinna Friedrich
- Institute of Reproductive Genetics, University of Münster, 48149, Münster, Germany
| | - Eric Bareke
- Department of Human Genetics, McGill University Health Centre, Montreal, QC, Canada
| | - Amira Nabil
- Department of Human Genetics, Medical Research Institute, Alexandria University, Alexandria, Egypt
| | - Frank Tüttelmann
- Institute of Reproductive Genetics, University of Münster, 48149, Münster, Germany
| | - Jacek Majewski
- Department of Human Genetics, McGill University Health Centre, Montreal, QC, Canada
| | - Ebtesam Abdalla
- Department of Human Genetics, Medical Research Institute, Alexandria University, Alexandria, Egypt
| | - Seang-Lin Tan
- OriginElle Fertility Clinic, Montreal, QC, Canada
- Department of Obstetrics and Gynecology, McGill University, Montreal, QC, Canada
| | - Rima Slim
- Department of Human Genetics, McGill University Health Centre, Montreal, QC, Canada.
- Department of Obstetrics and Gynecology, McGill University, Montreal, QC, Canada.
- Research Institute of the McGill University Health Centre, 1001 Décarie Blvd, Montréal, Québec, H4A 3J1, Canada.
| |
Collapse
|
30
|
Zhang J, Su T, Fan Y, Cheng C, Xu L, LiTian. Spotlight on iron overload and ferroptosis: Research progress in female infertility. Life Sci 2024; 340:122370. [PMID: 38141854 DOI: 10.1016/j.lfs.2023.122370] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Revised: 12/15/2023] [Accepted: 12/18/2023] [Indexed: 12/25/2023]
Abstract
Iron is an essential trace element for organisms. However, iron overload, which is common in haematological disorders (e.g. haemochromatosis, myelodysplastic syndromes, aplastic anaemia, and thalassaemia, blood transfusion-dependent or not), can promote reactive oxygen species generation and induce ferroptosis, a novel form of programmed cell death characterised by excess iron and lipid peroxidation, thus causing cell and tissue damage. Infertility is a global health concern. Recent evidence has indicated the emerging role of iron overload and ferroptosis in female infertility by inducing hypogonadism, causing ovary dysfunction, impairing preimplantation embryos, attenuating endometrial receptivity, and crosstalk between subfertility-related disorders, such as polycystic ovary syndrome and endometriosis. In addition, gut microbiota and their metabolites are involved in iron metabolism, ferroptosis, and female infertility. In this review, we systematically elaborate on the current research progress in female infertility with a novel focus on iron overload and ferroptosis and summarise promising therapies targeting iron overload and ferroptosis to recover fertility in women. In summary, our study provides new insights into female infertility and offers literature references for the clinical management of female infertility associated with iron overload and ferroptosis, which may be beneficial for females with haematopoietic disorders suffering from both iron overload and infertility.
Collapse
Affiliation(s)
- Jinghua Zhang
- Department of Obstetrics and Gynecology, Peking University People's Hospital, Beijing 100044, China; Reproductive Medical Center, Peking University People's Hospital, Beijing 100044, China
| | - Tiantian Su
- Department of Obstetrics and Gynecology, Peking University People's Hospital, Beijing 100044, China; Reproductive Medical Center, Peking University People's Hospital, Beijing 100044, China
| | - Yuan Fan
- Department of Obstetrics and Gynecology, Peking University People's Hospital, Beijing 100044, China; Reproductive Medical Center, Peking University People's Hospital, Beijing 100044, China
| | - Cheng Cheng
- Department of Obstetrics and Gynecology, Peking University People's Hospital, Beijing 100044, China; Reproductive Medical Center, Peking University People's Hospital, Beijing 100044, China
| | - Lanping Xu
- Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Peking University People's Hospital & Institute of Hematology, No. 11 Xizhimen South Street, Xicheng District, Beijing 100044, China
| | - LiTian
- Department of Obstetrics and Gynecology, Peking University People's Hospital, Beijing 100044, China; Reproductive Medical Center, Peking University People's Hospital, Beijing 100044, China.
| |
Collapse
|
31
|
Li L, Ding X, Sheft AP, Schimenti JC. A high throughput CRISPR perturbation screen identifies epigenetic regulators impacting primordial germ cell development. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.26.582097. [PMID: 38463983 PMCID: PMC10925113 DOI: 10.1101/2024.02.26.582097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/12/2024]
Abstract
Certain environmental factors can impact fertility and reproductive parameters such as the number and quality of sperm and eggs. One possible mechanism is the perturbation of epigenetic landscapes in the germline. To explore this possibility, we conducted a CRISPRi screen of epigenetic-related genes to identify those that specifically perturb the differentiation of embryonic stem cells (ESCs) into primordial germ cell-like cells (PGCLCs), exploiting a highly scalable cytokine-free platform. Of the 701 genes screened, inhibition of 53 decreased the efficiency of PGCLC formation. NCOR2, a transcriptional repressor that acts via recruitment of Class I and Class IIa histone deacetylases (HDACs) to gene targets, was particularly potent in suppressing PGCLC differentiation. Consistent with evidence that histone deacetylation is crucial for germline differentiation, we found that the HDAC inhibitors (HDACi) valproic acid (VPA; an anti-convulsant) and sodium butyrate (SB; a widely-used dietary supplement) also suppressed ESC>PGCLC differentiation. Furthermore, exposure of developing mouse embryos to SB or VPA caused hypospermatogenesis. Transcriptome analyses of HDACi-treated, differentiating ESC>PGCLC cultures revealed suppression of germline-associated pathways and enhancement of somatic pathways. This work demonstrates the feasibility of conducting large-scale functional screens of genes, chemicals, or other agents that may impact germline development.
Collapse
|
32
|
Sang L, Ge Y, Liu F, Wei K, Shen X, Zhang Y, Li Z, Lu W, Gao X, Zhang Y. Association between per- and polyfluoroalkyl substances and sex hormone levels in males based on human studies. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2024; 271:115998. [PMID: 38262091 DOI: 10.1016/j.ecoenv.2024.115998] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Revised: 01/13/2024] [Accepted: 01/17/2024] [Indexed: 01/25/2024]
Abstract
BACKGROUND Per- and poly-fluoroalkyl substances (PFAS) are ubiquitous chemicals in the environment and our daily lives. Several epidemiological studies have revealed that PFAS exposure is linked to male sex hormone levels; however, the conclusions are inconsistent across studies. Consequently, we performed a meta-analysis to systematically evaluate the association between PFAS exposure and male sex hormones. METHODS The Preferred Reporting Items for Systematic Reviews and Meta-Analysis (PRISMA) standards were followed during the meta-analysis. PubMed, Wed of Science, Embase, Cochrane Library, and Ovid databases were used to identify suitable articles before June 2023. The 95% CI and β values were calculated to assess the association between male sex hormone levels and PFAS exposure. Heterogeneity among the included studies was tested using inconsistency statistics (I2). RESULTS The literature search identified 12 published articles that met our search criteria, involving 7506 participants. Our results revealed that perfluorononanoic acid (PFNA) and perfluorooctanoic acid (PFOA) exposures were negatively correlated with testosterone (β = -0.05; 95% CI: -0.09, -0.02, P = 0.003) and (β = -0.04; 95% CI: -0.08, 0.00, P = 0.049), respectively. CONCLUSION Exposure to PFNA and PFOA is negatively correlated with changes in male testosterone levels. This correlation suggests that we need to pay attention in the future to whether they are potential risk factors for male reproductive health.
Collapse
Affiliation(s)
- Lingli Sang
- Department of Public Health, Kangda College of Nanjing Medical University, Lianyungang 222000, China
| | - Yue Ge
- Department of Public Health, Kangda College of Nanjing Medical University, Lianyungang 222000, China
| | - Fucun Liu
- Department of Public Health, Kangda College of Nanjing Medical University, Lianyungang 222000, China
| | - Kai Wei
- Department of Public Health, Kangda College of Nanjing Medical University, Lianyungang 222000, China
| | - Xingyu Shen
- Department of Public Health, Kangda College of Nanjing Medical University, Lianyungang 222000, China
| | - Yuxin Zhang
- Department of Public Health, Kangda College of Nanjing Medical University, Lianyungang 222000, China
| | - Zheng Li
- Department of Public Health, Kangda College of Nanjing Medical University, Lianyungang 222000, China
| | - Wencen Lu
- Department of Public Health, Kangda College of Nanjing Medical University, Lianyungang 222000, China
| | - Xia Gao
- Department of Otolaryngology Head and Neck Surgery of Xuzhou Cancer Hospital, XuZhou 2210000, China.
| | - Yan Zhang
- Department of Public Health, Kangda College of Nanjing Medical University, Lianyungang 222000, China.
| |
Collapse
|
33
|
Xue L, Wang S, Wei P, Liu H, Mao X, Qin J, Li Y, Zhang X, Li Z, Huang Y, Chen L, Shi W, Liu L. Early rescue oocyte activation at 5 h post-ICSI is a useful strategy for avoiding unexpected fertilization failure and low fertilization in ICSI cycles. Front Endocrinol (Lausanne) 2024; 14:1301505. [PMID: 38239979 PMCID: PMC10794723 DOI: 10.3389/fendo.2023.1301505] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Accepted: 12/12/2023] [Indexed: 01/22/2024] Open
Abstract
Introduction Attempts to artificially activate unfertilized oocytes at 24 h post intracytoplasmic sperm injection (ICSI) have generally resulted in poor outcomes. This study aims to explore a new strategy for early judgement and rescue activation of unfertilized oocytes at 5 h post ICSI to avoid unexpected fertilization failure (UFF) or unexpected low fertilization (ULF) in ICSI cycles. Methods Firstly, time-lapse data from 278 ICSI cycles were retrospectively analyzed to establish an indicator for fertilization failure prediction. Secondly, 14 UFF and 20 ULF cycles were enrolled for an observational study, early rescue oocyte activation (EROA) was performed on oocytes without post-ICSI Pb2 extrusion to investigate fertilization efficiency, embryo development and clinical outcomes. Results The average time to Pb2 extrusion post-ICSI was 3.03±1.21 h, 95.54% of oocytes had extruded Pb2 before 5 h, and the sensitivity and specificity for monitoring Pb2 extrusion at 5 h by time-lapse imaging to predict fertilization were 99.59% and 99.78%, respectively. Early rescue activation of oocytes with no Pb2 extrusion resulted in acceptable fertilization and embryo developmental outcomes, in terms of the fertilization rate (75.00, 72.99%), 2PN fertilization rate (61.36, 56.93%), good-quality embryo rate (42.59, 50.00%), blastocyst formation rate (48.28, 46.03%), good-quality blastocyst rate (34.48, 33.33%), and oocyte utilization rate (36.36, 27.74%), for both UFF and ULF cycles. The clinical pregnancy, embryo implantation, and early miscarriage rates in the rescue oocyte activation group did not significantly differ from those in the Pb2 extrusion group. Fourteen unexpected fertilization failures and 20 low fertilization ICSI cycles were rescued and resulted in clinical pregnancy rates of 40.00% (4/10) and 57.14% (8/14), respectively. Conclusions This study demonstrates that monitoring Pb2 extrusion by time-lapse imaging can accurately predict fertilization outcomes, suggesting that early rescue oocyte activation at 5 h post ICSI is an effective strategy for avoiding unexpected fertilization failure and low fertilization in ICSI cycles.
Collapse
Affiliation(s)
- Lintao Xue
- Reproductive Medical and Genetic Center, The People’s Hospital of GuangXi Zhuang Autonomous Region, Nanning, China
| | | | | | | | | | | | | | | | | | | | | | | | - Liling Liu
- Reproductive Medical and Genetic Center, The People’s Hospital of GuangXi Zhuang Autonomous Region, Nanning, China
| |
Collapse
|
34
|
Chen Y, Yang J, Zhang L. The Impact of Follicular Fluid Oxidative Stress Levels on the Outcomes of Assisted Reproductive Therapy. Antioxidants (Basel) 2023; 12:2117. [PMID: 38136236 PMCID: PMC10740420 DOI: 10.3390/antiox12122117] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Revised: 12/11/2023] [Accepted: 12/11/2023] [Indexed: 12/24/2023] Open
Abstract
Oocyte quality is a pivotal determinant of assisted reproductive outcomes. The quality of oocytes is intricately linked to their developmental microenvironment, particularly the levels of oxidative stress within the follicular fluid. Oxidative stress levels in follicular fluid may have a substantial influence on oocyte health, thereby impacting the outcomes of ART procedures. This review meticulously explores the intricate relationship between oxidative stress in follicular fluid and ART outcomes. Furthermore, it delves into strategies aimed at ameliorating the oxidative stress status of follicular fluid, with the overarching goal of enhancing the overall efficacy of ART. This research endeavors to establish a robust foundation and provide valuable guidance for clinical treatment approaches, particularly in the context of infertile women, including those of advanced maternal age.
Collapse
Affiliation(s)
| | | | - Ling Zhang
- Institute of Reproductive Health, Tongji Medical College, Huazhong University of Science and Technology, No. 13 Hangkong Road, Wuhan 430030, China; (Y.C.); (J.Y.)
| |
Collapse
|
35
|
Respekta N, Pich K, Mlyczyńska E, Dobrzyń K, Ramé C, Kamiński T, Smolińska N, Dupont J, Rak A. Plasma level of omentin-1, its expression, and its regulation by gonadotropin-releasing hormone and gonadotropins in porcine anterior pituitary cells. Sci Rep 2023; 13:19325. [PMID: 37935840 PMCID: PMC10630491 DOI: 10.1038/s41598-023-46742-4] [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: 08/18/2023] [Accepted: 11/04/2023] [Indexed: 11/09/2023] Open
Abstract
Omentin-1 (OMNT1) is an adipokine involved in the regulation of energy metabolism, insulin sensitivity, and reproduction. The present study was the first to investigate the plasma levels and expression of OMNT1 in the anterior pituitary (AP) gland on days 2-3, 10-12, 14-16, and 17-19 of the estrous cycle of normal-weight Large White (LW) and fat Meishan (MS) pigs. Next, we determined the effect of GnRH, LH, and FSH on the OMNT1 levels in cultured AP cells. The gene and protein expression of OMNT1 in AP fluctuated during the estrous cycle, with a higher expression in MS than in LW (except on days 10-12). However, plasma levels of OMNT1 were higher in LW than in MS. OMNT1 was localized in somatotrophs, lactotrophs, thyrotrophs, and gonadotrophs. In LW pituitary cells, GnRH and gonadotropins stimulated OMNT1 protein expression (except FSH on days 14-16) and had no effect on OMNT1 levels in the culture medium. In MS pituitary cells, we observed that GnRH and LH increased while FSH decreased OMNT1 protein expression. These findings showed OMNT1 expression and regulation in the porcine AP and suggested that OMNT1 could be a new player modifying the pituitary functions.
Collapse
Affiliation(s)
- Natalia Respekta
- Laboratory of Physiology and Toxicology of Reproduction, Institute of Zoology and Biomedical Research, Jagiellonian University, Gronostajowa 9 Street, 30-387, Kraków, Poland
- Doctoral School of Exact and Natural Sciences, Jagiellonian University, Kraków, Poland
| | - Karolina Pich
- Laboratory of Physiology and Toxicology of Reproduction, Institute of Zoology and Biomedical Research, Jagiellonian University, Gronostajowa 9 Street, 30-387, Kraków, Poland
- Doctoral School of Exact and Natural Sciences, Jagiellonian University, Kraków, Poland
| | - Ewa Mlyczyńska
- Laboratory of Physiology and Toxicology of Reproduction, Institute of Zoology and Biomedical Research, Jagiellonian University, Gronostajowa 9 Street, 30-387, Kraków, Poland
- Doctoral School of Exact and Natural Sciences, Jagiellonian University, Kraków, Poland
| | - Kamil Dobrzyń
- Department of Zoology, Faculty of Biology and Biotechnology, University of Warmia and Mazury, Kortowo, Olsztyn, Poland
| | - Christelle Ramé
- INRAE, UMR85, Unité Physiologie de la Reproduction et des Comportements, Nouzilly, France
| | - Tadeusz Kamiński
- Department of Animal Anatomy and Physiology, Faculty of Biology and Biotechnology, University of Warmia and Mazury, Kortowo, Olsztyn, Poland
| | - Nina Smolińska
- Department of Animal Anatomy and Physiology, Faculty of Biology and Biotechnology, University of Warmia and Mazury, Kortowo, Olsztyn, Poland
| | - Joëlle Dupont
- INRAE, UMR85, Unité Physiologie de la Reproduction et des Comportements, Nouzilly, France
| | - Agnieszka Rak
- Laboratory of Physiology and Toxicology of Reproduction, Institute of Zoology and Biomedical Research, Jagiellonian University, Gronostajowa 9 Street, 30-387, Kraków, Poland.
| |
Collapse
|
36
|
Tang M, Zhao M, Shi Y. New insight into the role of macrophages in ovarian function and ovarian aging. Front Endocrinol (Lausanne) 2023; 14:1282658. [PMID: 38027176 PMCID: PMC10662485 DOI: 10.3389/fendo.2023.1282658] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Accepted: 10/17/2023] [Indexed: 12/01/2023] Open
Abstract
Macrophages (MΦs) are the most abundant leukocytes in mammalian ovaries that have heterogeneity and plasticity. A body of evidence has indicated that these cells are important in maintaining ovarian homeostasis and they play critical roles in ovarian physiological events, such as folliculogenesis, ovulation, corpus luteum formation and regression. As females age, ovarian tissue microenvironment is typified by chronic inflammation with exacerbated ovarian fibrosis. In response to specific danger signals within aged ovaries, macrophages polarize into different M1 or M2 phenotypes, and specialize in unique functions to participate in the ovarian aging process. In this review, we will focus on the physiologic roles of MΦs in normal ovarian functions. Furthermore, we will discuss the roles of MΦs in the process of ovarian senescence, as well as the novel techniques applied in this field.
Collapse
Affiliation(s)
- Maoxing Tang
- Department of Reproductive Medicine, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Manzhi Zhao
- Department of Pulmonary and Critical Care Medicine, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Yuhua Shi
- Department of Reproductive Medicine, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| |
Collapse
|
37
|
Raoofi A, Omraninava M, Javan R, Maghsodi D, Rustamzadeh A, Nasiry D, Ghaemi A. Protective effects of epigallocatechin gallate in the mice induced by chronic scrotal hyperthermia. Tissue Cell 2023; 84:102165. [PMID: 37480630 DOI: 10.1016/j.tice.2023.102165] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Revised: 07/07/2023] [Accepted: 07/11/2023] [Indexed: 07/24/2023]
Abstract
One of the most common complications of chronic scrotal hyperthermia (SHT) is a serious disorder in the male reproductive system. The most important factor in the occurrence of these disorders is oxidative stress. Currently, we investigated the effects of epigallocatechin gallate (EGCG), as a highly potent antioxidant, against cells and tissue disorders in mice affected by chronic SHT. Fifty-six male adult NMRI mice were allocated into seven equal groups. Except the non-treated (Control) group, six other groups were exposed to heat stress. Two treated groups including Preventive and Curative received oral administration of EGCG (50 mg/kg/day) starting immediately before heat exposure and fifteen consecutive days after the end of the heat exposure, respectively. For each treated group, two subgroups including positive control (Pre/Cur + PC groups) and vehicle (Pre/Cur + vehicle groups) were considered. At the end of the study, sperm characteristics, testosterone levels, stereological parameters, apoptosis, oxidant state, and molecular assessments were performed. We found that the sperm parameters, testosterone levels, the numerical density of spermatogonia, primary spermatocytes, spermatids, sertoli, leydig cells, and seminiferous tubules, biochemical factors (except MDA), and expression of c-kit gene were significantly higher in the Preventive and Curative groups, especially in Preventive ones, compared to other groups (P < 0.05). This is while expression of HSP72 and NF-κβ genes, MDA levels, as well as density of apoptotic cells considerably decreased in both EGCG-treated groups compared to other groups and it was more pronounced in Preventive ones (P < 0.05). Generally, EGCG attenuated cellular and molecular disorders induced by heat stress in the testis and it was more pronounced in Preventive status.
Collapse
Affiliation(s)
- Amir Raoofi
- Cellular and Molecular Research Center, Sabzevar University of Medical Sciences, Sabzevar, Iran
| | - Melody Omraninava
- Health Reproductive Research Center, Islamic Azad University, Sari, Iran
| | - Roghayeh Javan
- Traditional and Complementary Medicine Research Center, Sabzevar University of Medical Science, Sabzevar, Iran
| | - Davood Maghsodi
- Student Research Committee, Sabzevar University of Medical Sciences, Sabzevar, Iran
| | - Auob Rustamzadeh
- Department of Anatomical sciences, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Davood Nasiry
- Department of Paramedicine, Amol School of Paramedical Sciences, Mazandaran University of Medical Sciences, Sari, Iran.
| | - Alireza Ghaemi
- Department of Basic Sciences and Nutrition, Health Sciences Research Center, Faculty of Public Health, Mazandaran University of Medical Sciences, Sari, Iran.
| |
Collapse
|
38
|
Martínez-Varea A, Martínez-Gómez M, Novillo B, Domenech J, Morales-Roselló J, Diago-Almela V. Perinatal Outcomes of Monochorionic Twin Pregnancies Conceived Naturally Versus through Assisted Reproductive Techniques. J Clin Med 2023; 12:6097. [PMID: 37763036 PMCID: PMC10531548 DOI: 10.3390/jcm12186097] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Revised: 09/09/2023] [Accepted: 09/20/2023] [Indexed: 09/29/2023] Open
Abstract
Objective: It has been reported that monochorionic twin pregnancies conceived through assisted reproductive techniques (ART) display a higher risk of second-trimester miscarriage, cesarean delivery, and neonatal death than those conceived naturally. The aim of this study was to compare the perinatal outcomes of monochorionic diamniotic (MCDA) twin pregnancies conceived naturally and through ART in a tertiary hospital. Methods: This was a retrospective cohort study of all MCDA twin pregnancies that received obstetric care and delivered at La Fe University and Polytechnic Hospital between 2015 and 2021. MCDA pregnancies that were referred to the tertiary hospital for specialized management, follow-up, and delivery were also included. The study was approved by The Health Research Institute Hospital La Fe (IIS La Fe). Results: Among the 184 MCDA pregnancies, 149 (81%) had a natural conception, and 35 (19%) were conceived through ART. Patients with an MCDA pregnancy who conceived through ART had a significantly older maternal age (38.0 [35.5-42.5] vs. 32.0 [29.0-36.0], p < 0.001) and an elevated rate of nulliparity (80.0% vs. 50.3%, p = 0.001). Regarding pregnancy complications, MCDA pregnancies through ART were associated with a significantly higher incidence of gestational diabetes (22.9% vs. 2.7%, p < 0.001), hypertensive disorders during pregnancy (22.9% vs. 9.4%, p = 0.04), and other pregnancy complications such as threatened labor or preterm prelabor rupture of membranes (14.3% vs. 36.2%, p = 0.015), than naturally conceived MCDA pregnancies. No differences were found in the incidence of twin-to-twin transfusion syndrome (20% vs. 33.6%, p = 0.155). MCDA pregnancies through natural conception had a greater rate of vaginal delivery than MCDA through ART (16.8% vs. 2.9%, p = 0.032). When adjusted for confounding factors, MCDA pregnancies through ART were only more likely to develop gestational diabetes than those naturally conceived (aOR 7.86, 95% CI 1.55-39.87). No differences were found regarding neonatal outcomes between groups. Conclusions: Compared with naturally conceived MCDA twin pregnancies, those conceived through ART displayed a significantly higher risk of developing gestational diabetes. No differences regarding other pregnancy complications, mode of delivery, or neonatal outcomes were found between groups.
Collapse
Affiliation(s)
- Alicia Martínez-Varea
- Department of Obstetrics and Gynaecology, La Fe University and Polytechnic Hospital, Avenida Fernando Abril Martorell 106, 46026 Valencia, Spain; (M.M.-G.); (B.N.); (J.M.-R.); (V.D.-A.)
| | - Martha Martínez-Gómez
- Department of Obstetrics and Gynaecology, La Fe University and Polytechnic Hospital, Avenida Fernando Abril Martorell 106, 46026 Valencia, Spain; (M.M.-G.); (B.N.); (J.M.-R.); (V.D.-A.)
| | - Blanca Novillo
- Department of Obstetrics and Gynaecology, La Fe University and Polytechnic Hospital, Avenida Fernando Abril Martorell 106, 46026 Valencia, Spain; (M.M.-G.); (B.N.); (J.M.-R.); (V.D.-A.)
| | - Josep Domenech
- Department of Economics and Social Sciences, Universitat Politècnica de València, Camí de Vera s/n, 46022 Valencia, Spain;
| | - José Morales-Roselló
- Department of Obstetrics and Gynaecology, La Fe University and Polytechnic Hospital, Avenida Fernando Abril Martorell 106, 46026 Valencia, Spain; (M.M.-G.); (B.N.); (J.M.-R.); (V.D.-A.)
- Department of Pediatrics, Obstetrics and Gynecology, University of Valencia, Avenida Blasco Ibáñez 15, 46010 Valencia, Spain
| | - Vicente Diago-Almela
- Department of Obstetrics and Gynaecology, La Fe University and Polytechnic Hospital, Avenida Fernando Abril Martorell 106, 46026 Valencia, Spain; (M.M.-G.); (B.N.); (J.M.-R.); (V.D.-A.)
| |
Collapse
|
39
|
Yang J, Tang J, He X, Di R, Zhang X, Zhang J, Guo X, Chu M, Hu W. Comparative Transcriptomics Identify Key Pituitary Circular RNAs That Participate in Sheep ( Ovis aries) Reproduction. Animals (Basel) 2023; 13:2711. [PMID: 37684975 PMCID: PMC10486758 DOI: 10.3390/ani13172711] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Revised: 08/15/2023] [Accepted: 08/17/2023] [Indexed: 09/10/2023] Open
Abstract
CircRNAs have been found to play key roles in many biological processes and have diverse biological functions. There have been studies on circRNAs in sheep pituitary, and some important circRNAs have been found. But there are still few studies on circRNAs in sheep pituitary with different fecundity. In this study, we obtained the circRNAs expression profiles in the pituitary of FecB ++ genotype Small Tail Han sheep with different fecundity and estrous phases. A total of 34,878 circRNAs were identified in 12 pituitary samples, 300 differentially expressed circRNAs (DE circRNAs) (down: 104; up: 196) were identified in polytocous sheep in the follicular phase (PF) and monotocous sheep in the follicular phase (MF) (PF vs. MF), and 347 DE circRNAs (down: 162; up: 185) were identified in polytocous sheep in the luteal phase (PL) and monotocous sheep in the luteal phase (ML) (PL vs. ML). Cortisol synthesis and secretion pathway (follicular phase) and estrogen signaling pathway (luteal phase) were obtained by functional enrichment analysis of circRNAs source genes. Competing endogenous RNA (ceRNA) network analysis of key DE circRNAs revealed that oar-circ-0022776 (source gene ITPR2, follicular phase) targeted oar-miR-432, oar-circ-0009003 (source gene ITPR1, luteal phase) and oar-circ-0003113 (source gene PLCB1, luteal phase) targeted oar-miR-370-3p. We also explored the coding ability of DE circRNAs. In conclusion, our study shows that changes in the pituitary circRNAs may be related to the response of the pituitary to steroid hormones and regulate the reproductive process of sheep by affecting the pituitary function. Results of this study provide some new information for understanding the functions of circRNAs and the fecundity of FecB ++ genotype sheep.
Collapse
Affiliation(s)
- Jianqi Yang
- State Key Laboratory of Animal Biotech Breeding, Institute of Animal Science, Chinese Academy of Agricultural Sciences (CAAS), Beijing 100193, China; (J.Y.); (X.H.); (R.D.)
| | - Jishun Tang
- Institute of Animal Husbandry and Veterinary Medicine, Anhui Academy of Agricultural Sciences, Hefei 230031, China;
| | - Xiaoyun He
- State Key Laboratory of Animal Biotech Breeding, Institute of Animal Science, Chinese Academy of Agricultural Sciences (CAAS), Beijing 100193, China; (J.Y.); (X.H.); (R.D.)
| | - Ran Di
- State Key Laboratory of Animal Biotech Breeding, Institute of Animal Science, Chinese Academy of Agricultural Sciences (CAAS), Beijing 100193, China; (J.Y.); (X.H.); (R.D.)
| | - Xiaosheng Zhang
- Tianjin Key Laboratory of Animal Molecular Breeding and Biotechnology, Tianjin Engineering Research Center of Animal Healthy Farming, Institute of Animal Science and Veterinary, Tianjin Academy of Agricultural Sciences, Tianjin 300381, China; (X.Z.); (J.Z.); (X.G.)
| | - Jinlong Zhang
- Tianjin Key Laboratory of Animal Molecular Breeding and Biotechnology, Tianjin Engineering Research Center of Animal Healthy Farming, Institute of Animal Science and Veterinary, Tianjin Academy of Agricultural Sciences, Tianjin 300381, China; (X.Z.); (J.Z.); (X.G.)
| | - Xiaofei Guo
- Tianjin Key Laboratory of Animal Molecular Breeding and Biotechnology, Tianjin Engineering Research Center of Animal Healthy Farming, Institute of Animal Science and Veterinary, Tianjin Academy of Agricultural Sciences, Tianjin 300381, China; (X.Z.); (J.Z.); (X.G.)
| | - Mingxing Chu
- State Key Laboratory of Animal Biotech Breeding, Institute of Animal Science, Chinese Academy of Agricultural Sciences (CAAS), Beijing 100193, China; (J.Y.); (X.H.); (R.D.)
| | - Wenping Hu
- State Key Laboratory of Animal Biotech Breeding, Institute of Animal Science, Chinese Academy of Agricultural Sciences (CAAS), Beijing 100193, China; (J.Y.); (X.H.); (R.D.)
| |
Collapse
|
40
|
Albertini DF. Continuing the search for sperm-based determinants of early mammalian development. J Assist Reprod Genet 2023; 40:695-696. [PMID: 37199868 PMCID: PMC10224872 DOI: 10.1007/s10815-023-02826-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/19/2023] Open
|