1
|
Dhyani K, Dash S, Joshi S, Garg A, Pal D, Nishant K, Muniyappa K. The ATPase activity of yeast chromosome axis protein Hop1 affects the frequency of meiotic crossovers. Nucleic Acids Res 2025; 53:gkae1264. [PMID: 39727188 PMCID: PMC11797056 DOI: 10.1093/nar/gkae1264] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Revised: 12/05/2024] [Accepted: 12/10/2024] [Indexed: 12/28/2024] Open
Abstract
Saccharomyces cerevisiae meiosis-specific Hop1, a structural constituent of the synaptonemal complex, also facilitates the formation of programmed DNA double-strand breaks and the pairing of homologous chromosomes. Here, we reveal a serendipitous discovery that Hop1 possesses robust DNA-independent ATPase activity, although it lacks recognizable sequence motifs required for ATP binding and hydrolysis. By leveraging molecular docking combined with molecular dynamics simulations and biochemical assays, we identified an ensemble of five amino acid residues in Hop1 that could potentially participate in ATP-binding and hydrolysis. Consistent with this premise, we found that Hop1 binds to ATP and that substitution of amino acid residues in the putative ATP-binding site significantly impaired its ATPase activity, suggesting that this activity is intrinsic to Hop1. Notably, K65A and N67Q substitutions in the Hop1 N-terminal HORMA domain synergistically abolished its ATPase activity, noticeably impaired its DNA-binding affinity and reduced its association with meiotic chromosomes, while enhancing the frequency of meiotic crossovers (COs). Overall, our study establishes Hop1 as a DNA-independent ATPase and reveals a potential biological function for its ATPase activity in the regulation of meiotic CO frequency.
Collapse
Affiliation(s)
- Kshitiza M Dhyani
- Department of Biochemistry, Indian Institute of Science, CV Raman Road, Bengaluru 560012, India
| | - Suman Dash
- School of Biology, Indian Institute of Science Education and Research, Maruthamala(PO), Vithura, Thiruvananthapuram 695551, India
| | - Sameer Joshi
- School of Biology, Indian Institute of Science Education and Research, Maruthamala(PO), Vithura, Thiruvananthapuram 695551, India
| | - Aditi Garg
- Computational and Data Sciences, Indian Institute of Science, CV Raman Road, Bengaluru 560012, India
| | - Debnath Pal
- Computational and Data Sciences, Indian Institute of Science, CV Raman Road, Bengaluru 560012, India
| | - Koodali T Nishant
- School of Biology, Indian Institute of Science Education and Research, Maruthamala(PO), Vithura, Thiruvananthapuram 695551, India
| | - Kalappa Muniyappa
- Department of Biochemistry, Indian Institute of Science, CV Raman Road, Bengaluru 560012, India
| |
Collapse
|
2
|
Ito M, Yun Y, Kulkarni DS, Lee S, Sandhu S, Nuñez B, Hu L, Lee K, Lim N, Hirota RM, Prendergast R, Huang C, Huang I, Hunter N. Distinct and interdependent functions of three RING proteins regulate recombination during mammalian meiosis. Proc Natl Acad Sci U S A 2025; 122:e2412961121. [PMID: 39761402 PMCID: PMC11745341 DOI: 10.1073/pnas.2412961121] [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] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Accepted: 10/21/2024] [Indexed: 01/30/2025] Open
Abstract
During meiosis, each pair of homologous chromosomes becomes connected by at least one crossover, as required for accurate segregation, and adjacent crossovers are widely separated thereby limiting total numbers. In coarsening models, this crossover patterning results from nascent recombination sites competing to accrue a limiting pro-crossover RING-domain protein (COR) that diffuses between synapsed chromosomes. Here, we delineate the localization dynamics of three mammalian CORs in the mouse and determine their interdependencies. RNF212, HEI10, and the newest member RNF212B show divergent spatiotemporal dynamics along synapsed chromosomes, including profound differences in spermatocytes and oocytes, that are not easily reconciled by elementary coarsening models. Contrasting mutant phenotypes and genetic requirements indicate that RNF212B, RNF212, and HEI10 play distinct but interdependent functions in regulating meiotic recombination and coordinating the events of meiotic prophase-I by integrating signals from DNA breaks, homolog synapsis, the cell-cycle, and incipient crossover sites.
Collapse
Affiliation(s)
- Masaru Ito
- HHMI, University of California, Davis, CA95616
- Department of Microbiology & Molecular Genetics, University of California, Davis, CA95616
- Institute for Protein Research, Osaka University, Osaka565-0871, Japan
| | - Yan Yun
- HHMI, University of California, Davis, CA95616
- Department of Microbiology & Molecular Genetics, University of California, Davis, CA95616
- Center for Reproductive Medicine, Clinical Research Center, Shantou Central Hospital, Shantou, China515041
| | - Dhananjaya S. Kulkarni
- HHMI, University of California, Davis, CA95616
- Department of Microbiology & Molecular Genetics, University of California, Davis, CA95616
| | - Sunkyung Lee
- HHMI, University of California, Davis, CA95616
- Department of Microbiology & Molecular Genetics, University of California, Davis, CA95616
| | - Sumit Sandhu
- HHMI, University of California, Davis, CA95616
- Department of Microbiology & Molecular Genetics, University of California, Davis, CA95616
| | - Briana Nuñez
- HHMI, University of California, Davis, CA95616
- Department of Biochemistry & Molecular Biology, Brown University, Providence, RI02912
| | - Linya Hu
- Department of Microbiology & Molecular Genetics, University of California, Davis, CA95616
| | - Kevin Lee
- Department of Microbiology & Molecular Genetics, University of California, Davis, CA95616
| | - Nelly Lim
- Department of Microbiology & Molecular Genetics, University of California, Davis, CA95616
| | - Rachel M. Hirota
- Department of Microbiology & Molecular Genetics, University of California, Davis, CA95616
| | - Rowan Prendergast
- Department of Microbiology & Molecular Genetics, University of California, Davis, CA95616
| | - Cynthia Huang
- Department of Microbiology & Molecular Genetics, University of California, Davis, CA95616
| | - Ivy Huang
- Department of Microbiology & Molecular Genetics, University of California, Davis, CA95616
| | - Neil Hunter
- HHMI, University of California, Davis, CA95616
- Department of Microbiology & Molecular Genetics, University of California, Davis, CA95616
- Department of Molecular & Cellular Biology, University of California, Davis, CA95616
| |
Collapse
|
3
|
Topaloudis A, Cumer T, Lavanchy E, Ducrest AL, Simon C, Machado AP, Paposhvili N, Roulin A, Goudet J. The recombination landscape of the barn owl, from families to populations. Genetics 2025; 229:1-50. [PMID: 39545468 PMCID: PMC11708917 DOI: 10.1093/genetics/iyae190] [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: 07/26/2024] [Accepted: 11/01/2024] [Indexed: 11/17/2024] Open
Abstract
Homologous recombination is a meiotic process that generates diversity along the genome and interacts with all evolutionary forces. Despite its importance, studies of recombination landscapes are lacking due to methodological limitations and limited data. Frequently used approaches include linkage mapping based on familial data that provides sex-specific broad-scale estimates of realized recombination and inferences based on population linkage disequilibrium that reveal a more fine-scale resolution of the recombination landscape, albeit dependent on the effective population size and the selective forces acting on the population. In this study, we use a combination of these 2 methods to elucidate the recombination landscape for the Afro-European barn owl (Tyto alba). We find subtle differences in crossover placement between sexes that lead to differential effective shuffling of alleles. Linkage disequilibrium-based estimates of recombination are concordant with family-based estimates and identify large variation in recombination rates within and among linkage groups. Larger chromosomes show variation in recombination rates, while smaller chromosomes have a universally high rate that shapes the diversity landscape. We find that recombination rates are correlated with gene content, genetic diversity, and GC content. We find no conclusive differences in the recombination landscapes between populations. Overall, this comprehensive analysis enhances our understanding of recombination dynamics, genomic architecture, and sex-specific variation in the barn owl, contributing valuable insights to the broader field of avian genomics.
Collapse
Affiliation(s)
- Alexandros Topaloudis
- Department of Ecology and Evolution, University of Lausanne, Lausanne 1015, Switzerland
- Swiss Institute of Bioinformatics, Lausanne 1015, Switzerland
| | - Tristan Cumer
- Department of Ecology and Evolution, University of Lausanne, Lausanne 1015, Switzerland
- Swiss Institute of Bioinformatics, Lausanne 1015, Switzerland
| | - Eléonore Lavanchy
- Department of Ecology and Evolution, University of Lausanne, Lausanne 1015, Switzerland
- Swiss Institute of Bioinformatics, Lausanne 1015, Switzerland
| | - Anne-Lyse Ducrest
- Department of Ecology and Evolution, University of Lausanne, Lausanne 1015, Switzerland
| | - Celine Simon
- Department of Ecology and Evolution, University of Lausanne, Lausanne 1015, Switzerland
| | - Ana Paula Machado
- Department of Ecology and Evolution, University of Lausanne, Lausanne 1015, Switzerland
| | - Nika Paposhvili
- Institute of Ecology, Ilia State University, Tbilisi 0162, Georgia
| | - Alexandre Roulin
- Department of Ecology and Evolution, University of Lausanne, Lausanne 1015, Switzerland
| | - Jérôme Goudet
- Department of Ecology and Evolution, University of Lausanne, Lausanne 1015, Switzerland
- Swiss Institute of Bioinformatics, Lausanne 1015, Switzerland
| |
Collapse
|
4
|
Parée T, Noble L, Roze D, Teotónio H. Selection Can Favor a Recombination Landscape That Limits Polygenic Adaptation. Mol Biol Evol 2025; 42:msae273. [PMID: 39776196 PMCID: PMC11739800 DOI: 10.1093/molbev/msae273] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2024] [Revised: 12/05/2024] [Accepted: 12/13/2024] [Indexed: 01/11/2025] Open
Abstract
Modifiers of recombination rates have been described but the selective pressures acting on them and their effect on adaptation to novel environments remain unclear. We performed experimental evolution in the nematode Caenorhabditis elegans using alternative rec-1 alleles modifying the position of meiotic crossovers along chromosomes without detectable direct fitness effects. We show that adaptation to a novel environment is impaired by the allele that decreases recombination rates in the genomic regions containing fitness variation. However, the allele that impairs adaptation is indirectly favored by selection, because it increases recombination rates and reduces the associations among beneficial and deleterious variation located in its chromosomal vicinity. These results validate theoretical expectations about the evolution of recombination but suggest that genome-wide polygenic adaptation is of little consequence to indirect selection on recombination rate modifiers.
Collapse
Affiliation(s)
- Tom Parée
- Institut de Biologie, École Normale Supérieure, CNRS UMR 8197, Inserm U1024, PSL Research University, Paris 75005, France
- Department of Biology, New York University, New York, NY 10003, USA
| | - Luke Noble
- Institut de Biologie, École Normale Supérieure, CNRS UMR 8197, Inserm U1024, PSL Research University, Paris 75005, France
- EnviroDNA, 95 Albert St Brunswick, Melbourne, Victoria 3065, Australia
| | - Denis Roze
- Adaptation et Diversité en Milieu Marin CNRS UMR 7144, Station Biologique de Roscoff, Sorbonne University, Roscoff 29688, France
| | - Henrique Teotónio
- Institut de Biologie, École Normale Supérieure, CNRS UMR 8197, Inserm U1024, PSL Research University, Paris 75005, France
| |
Collapse
|
5
|
Han Y, Du Z, Wu H, Zhao R, Liu J, Gao S, Zeng S. CALB1 and RPL23 Are Essential for Maintaining Oocyte Quality and Function During Aging. Aging Cell 2025:e14466. [PMID: 39748132 DOI: 10.1111/acel.14466] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2024] [Revised: 12/04/2024] [Accepted: 12/12/2024] [Indexed: 01/04/2025] Open
Abstract
With advancing age, significant changes occur in the female reproductive system, the most notable of which is the decline in oocyte quality, a key factor affecting female fertility. However, the mechanisms underlying oocyte aging remain poorly understood. In this study, we obtained oocytes from aged and young female mice and performed single-cell transcriptome sequencing, comparing our findings with existing proteomic analyses. Our analysis revealed that one of the primary characteristics of aging oocytes is the disruption of calcium ion homeostasis. Specifically, we identified two key genes involved in the oocyte aging process, Calb1 and Rpl23. Experimental validation demonstrated that knockdown of CALB1 in oocytes led to reduced calcium ion levels in the endoplasmic reticulum and mitochondria, resulting in mitochondrial dysfunction and meiotic defects. Further experiments suggested that RPL23 may function as a downstream gene of CALB1, and its knockdown caused mitochondrial dysfunction, excessive accumulation of reactive oxygen species (ROS), and spindle assembly defects. Notably, overexpression of these two genes in aging oocytes partially rescued the maternal age-related defective phenotypes, underscoring their crucial roles in oocyte aging. This study provides a comprehensive understanding of the specific mechanisms underlying mouse oocyte aging at single-cell resolution, supported by experimental validation, and offers new directions and potential targets for future research into age-related reproductive health issues.
Collapse
Affiliation(s)
- Yingxue Han
- State Key Laboratory of Animal Biotech Breeding, National Engineering Laboratory for Animal Breeding, Key Laboratory of Animal Genetics, Breeding and Reproduction of the Ministry of Agriculture, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Zihuan Du
- National key Laboratory of Animal Breeding, College of Animal Science and Technology, Frontiers Science Center for Molecular Design Breeding (MOE), China Agricultural University, Beijing, China
| | - Hao Wu
- State Key Laboratory of Animal Biotech Breeding, National Engineering Laboratory for Animal Breeding, Key Laboratory of Animal Genetics, Breeding and Reproduction of the Ministry of Agriculture, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Rong Zhao
- State Key Laboratory of Animal Biotech Breeding, National Engineering Laboratory for Animal Breeding, Key Laboratory of Animal Genetics, Breeding and Reproduction of the Ministry of Agriculture, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Jikang Liu
- State Key Laboratory of Animal Biotech Breeding, National Engineering Laboratory for Animal Breeding, Key Laboratory of Animal Genetics, Breeding and Reproduction of the Ministry of Agriculture, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Shuai Gao
- National key Laboratory of Animal Breeding, College of Animal Science and Technology, Frontiers Science Center for Molecular Design Breeding (MOE), China Agricultural University, Beijing, China
| | - Shenming Zeng
- State Key Laboratory of Animal Biotech Breeding, National Engineering Laboratory for Animal Breeding, Key Laboratory of Animal Genetics, Breeding and Reproduction of the Ministry of Agriculture, College of Animal Science and Technology, China Agricultural University, Beijing, China
| |
Collapse
|
6
|
Varshavsky JR, Lam J, Cooper C, Allard P, Fung J, Oke A, Kumar R, Robinson JF, Woodruff TJ. Analyzing high-throughput assay data to advance the rapid screening of environmental chemicals for human reproductive toxicity. Reprod Toxicol 2025; 131:108725. [PMID: 39419704 DOI: 10.1016/j.reprotox.2024.108725] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Revised: 08/28/2024] [Accepted: 09/22/2024] [Indexed: 10/19/2024]
Abstract
While high-throughput (HTP) assays have been proposed as platforms to rapidly assess reproductive toxicity, there is currently a lack of established assays that specifically address germline development/function and fertility. We assessed the applicability domains of yeast (S. cerevisiae) and nematode (C. elegans) HTP assays in toxicity screening of 124 environmental chemicals, determining their agreement in identifying toxicants and their concordance with reproductive toxicity in vivo. We integrated data generated in the two models and compared results using a streamlined, semi-automated benchmark dose (BMD) modeling approach. We then extracted and modeled relevant mammalian in vivo data available for the matching chemicals included in the Toxicological Reference Database (ToxRefDB). We ranked potencies of common compounds using the BMD and evaluated correlation between the datasets using Pearson and Spearman correlation coefficients. We found moderate to good correlation across the three data sets, with r = 0.48 (95 % CI: 0.28-1.00, p<0.001) and rs = 0.40 (p=0.002) for the parametric and rank order correlations between the HTP BMDs; r = 0.95 (95 % CI: 0.76-1.00, p=0.0005) and rs = 0.89 (p=0.006) between the yeast assay and ToxRefDB BMDs; and r = 0.81 (95 % CI: 0.28-1.00, p=0.014) and rs = 0.75 (p=0.033) between the worm assay and ToxRefDB BMDs. Our findings underscore the potential of these HTP assays to identify environmental chemicals that exhibit reproductive toxicity. Integrating these HTP datasets into mammalian in vivo prediction models using machine learning methods could further enhance their predictive value in future rapid screening efforts.
Collapse
Affiliation(s)
- Julia R Varshavsky
- Department of Public Health and Health Sciences and Department of Civil and Environmental Engineering, Northeastern University, Boston, MA, USA.
| | - Juleen Lam
- Department of Public Health, California State University, East Bay, Hayward, CA, USA
| | - Courtney Cooper
- University of California, San Francisco (UCSF), Program on Reproductive Health and the Environment, Department of Obstetrics, Gynecology and Reproductive Sciences, San Francisco, CA, USA
| | - Patrick Allard
- Institute for Society and Genetics, University of California, Los Angeles (UCLA), Los Angeles, CA, USA
| | - Jennifer Fung
- Center for Reproductive Sciences and Department of Obstetrics, Gynecology & Reproductive Sciences, UCSF, San Francisco, CA, USA
| | - Ashwini Oke
- Center for Reproductive Sciences and Department of Obstetrics, Gynecology & Reproductive Sciences, UCSF, San Francisco, CA, USA
| | - Ravinder Kumar
- Center for Reproductive Sciences and Department of Obstetrics, Gynecology & Reproductive Sciences, UCSF, San Francisco, CA, USA
| | - Joshua F Robinson
- University of California, San Francisco (UCSF), Program on Reproductive Health and the Environment, Department of Obstetrics, Gynecology and Reproductive Sciences, San Francisco, CA, USA; Center for Reproductive Sciences and Department of Obstetrics, Gynecology & Reproductive Sciences, UCSF, San Francisco, CA, USA
| | - Tracey J Woodruff
- University of California, San Francisco (UCSF), Program on Reproductive Health and the Environment, Department of Obstetrics, Gynecology and Reproductive Sciences, San Francisco, CA, USA.
| |
Collapse
|
7
|
Payero L, Alani E. Crossover recombination between homologous chromosomes in meiosis: recent progress and remaining mysteries. Trends Genet 2025; 41:47-59. [PMID: 39490337 DOI: 10.1016/j.tig.2024.09.009] [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: 08/04/2024] [Revised: 09/26/2024] [Accepted: 09/27/2024] [Indexed: 11/05/2024]
Abstract
Crossing over between homologous chromosomes in meiosis is essential in most eukaryotes to produce gametes with the correct ploidy. Meiotic crossovers are typically evenly spaced, with each homolog pair receiving at least one crossover. The association of crossovers with distal sister chromatid cohesion is critical for the proper segregation of homologs in the first meiotic division. Studies in baker's yeast (Saccharomyces cerevisiae) have shown that meiotic crossovers result primarily from the biased resolution of double Holliday junction (dHJ) recombination intermediates through the actions of factors that belong to the DNA mismatch repair family. These findings and studies involving fine-scale mapping of meiotic crossover events have led to a new generation of mechanistic models for crossing over that are currently being tested.
Collapse
Affiliation(s)
- Lisette Payero
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, NY, USA
| | - Eric Alani
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, NY, USA.
| |
Collapse
|
8
|
Wedenoja S, Pihlajamäki M, Gissler M, Wedenoja J, Öhman H, Heinonen S, Kere J, Kääriäinen H, Tanner L. Infertility following trisomic pregnancies: A nationwide cohort study. Int J Gynaecol Obstet 2025; 168:326-332. [PMID: 39056516 PMCID: PMC11649879 DOI: 10.1002/ijgo.15828] [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: 06/06/2024] [Revised: 07/14/2024] [Accepted: 07/15/2024] [Indexed: 07/28/2024]
Abstract
OBJECTIVE To study whether gynecologic or reproductive disorders show association with trisomic conceptions. METHODS This nationwide cohort study utilized the Registry of Congenital Malformations to identify women who had a trisomic pregnancy (n = 5784), either with trisomy 13 (T13; n = 351), trisomy 18 (T18; n = 1065) or trisomy 21 (T21; n = 4369) from 1987 to 2018. We used the Finnish Maternity cohort to match the cases to population controls (n = 34 422) on the age, residence, and timing of pregnancy. These data were cross-linked to the ICD-10 diagnoses of the national Care Registry for Health Care data on specialized health care in Finland during 1996 to 2019. Both inflammatory (ICD-10 diagnoses: N70-N77) and noninflammatory disorders of the genital tract (N80-N98) were studied. Crude odds ratios (ORs) with 95% CIs were calculated for associations between diagnoses and trisomic conceptions. RESULTS The diagnosis of female infertility (N97) at any time was associated with trisomic conceptions (OR: 1.19, 95% CI: 1.08-1.32). In the subgroup analysis, this association was found for T18 (OR: 1.29, 95% CI: 1.03-1.61) and T21 (OR: 1.17, 95% CI: 1.04-1.32), but not for T13 (OR: 1.15, 95% CI: 0.75-1.72). When restricting the timing of the diagnosis of female infertility, an elevated OR was found only after the index pregnancy (OR: 1.81, 95% CI: 1.56-2.09). These increased odds for infertility after trisomic conceptions were observed both in women <35 years (T18 OR: 1.91, 95% CI: 1.21-3.00; T21 OR: 1.68, 95% CI: 1.31-2.14) and in women ≥35 years (T18 OR: 2.17, 95% CI: 1.40-3.33; T21 OR: 1.87; 95% CI: 1.47-2.39), but not after T13 conceptions. CONCLUSION Our observational data suggest a link between trisomic conceptions and subsequent diagnoses of infertility but do not demonstrate causality. These data implicate that partially similar mechanisms might predispose to trisomy and infertility, regardless of maternal age.
Collapse
Affiliation(s)
- Satu Wedenoja
- Information BrokersFinnish Institute for Health and WelfareHelsinkiFinland
- Obstetrics and GynecologyUniversity of Helsinki and Helsinki University HospitalHelsinkiFinland
- Stem Cells and Metabolism Research ProgramUniversity of HelsinkiHelsinkiFinland
- Folkhälsan Research CenterHelsinkiFinland
| | - Mika Pihlajamäki
- Information BrokersFinnish Institute for Health and WelfareHelsinkiFinland
| | - Mika Gissler
- Information BrokersFinnish Institute for Health and WelfareHelsinkiFinland
- Research Center for Child PsychiatryUniversity of TurkuTurkuFinland
- Region StockholmAcademic Primary Health Care CenterStockholmSweden
- Karolinska InstitutetDepartment of Molecular Medicine and SurgeryStockholmSweden
| | - Juho Wedenoja
- Department of OphthalmologyUniversity of Helsinki and Helsinki University HospitalHelsinkiFinland
| | - Hanna Öhman
- Biobank Borealis of Northern FinlandOulu University HospitalOuluFinland
- Faculty of MedicineUniversity of OuluOuluFinland
| | - Seppo Heinonen
- Obstetrics and GynecologyUniversity of Helsinki and Helsinki University HospitalHelsinkiFinland
| | - Juha Kere
- Stem Cells and Metabolism Research ProgramUniversity of HelsinkiHelsinkiFinland
- Folkhälsan Research CenterHelsinkiFinland
- Department of Biosciences and NutritionKarolinska InstitutetHuddingeSweden
| | - Helena Kääriäinen
- Genomics and Biomarkers UnitNational Institute for Health and WelfareHelsinkiFinland
| | - Laura Tanner
- Department of Clinical GeneticsHelsinki University HospitalHelsinkiFinland
- Department of Medical and Clinical GeneticsUniversity of HelsinkiHelsinkiFinland
| |
Collapse
|
9
|
Patel B, Grobler M, Herrera A, Logari E, Ortiz V, Bhalla N. The conserved ATPase PCH-2 controls the number and distribution of crossovers by antagonizing their formation in C. elegans. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.13.607819. [PMID: 39185160 PMCID: PMC11343117 DOI: 10.1101/2024.08.13.607819] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 08/27/2024]
Abstract
Meiotic crossover recombination is essential for both accurate chromosome segregation and the generation of new haplotypes for natural selection to act upon. This requirement is known as crossover assurance and is one example of crossover control. While the conserved role of the ATPase, PCH-2, during meiotic prophase has been enigmatic, a universal phenotype when pch-2 or its orthologs are mutated is a change in the number and distribution of meiotic crossovers. Here, we show that PCH-2 controls the number and distribution of crossovers by antagonizing their formation. This antagonism produces different effects at different stages of meiotic prophase: early in meiotic prophase, PCH-2 prevents double strand breaks from becoming crossover-eligible intermediates, limiting crossover formation at sites of initial double strand break formation and homolog interactions. Later in meiotic prophase, PCH-2 winnows the number of crossover-eligible intermediates, contributing to the designation of crossovers and ultimately, crossover assurance. We also demonstrate that PCH-2 accomplishes this regulation through the meiotic HORMAD, HIM-3. Our data strongly support a model in which PCH-2's conserved role is to remodel meiotic HORMADs throughout meiotic prophase to destabilize crossover-eligible precursors, coordinate meiotic recombination with synapsis, and contribute to the progressive implementation of meiotic recombination, guaranteeing crossover control.
Collapse
Affiliation(s)
- Bhumil Patel
- Department of Molecular, Cell and Developmental Biology, University of California, Santa Cruz, Santa Cruz, CA 95064
| | - Maryke Grobler
- Department of Molecular, Cell and Developmental Biology, University of California, Santa Cruz, Santa Cruz, CA 95064
| | - Alberto Herrera
- Department of Molecular, Cell and Developmental Biology, University of California, Santa Cruz, Santa Cruz, CA 95064
| | - Elias Logari
- Department of Molecular, Cell and Developmental Biology, University of California, Santa Cruz, Santa Cruz, CA 95064
| | - Valery Ortiz
- Department of Molecular, Cell and Developmental Biology, University of California, Santa Cruz, Santa Cruz, CA 95064
| | - Needhi Bhalla
- Department of Molecular, Cell and Developmental Biology, University of California, Santa Cruz, Santa Cruz, CA 95064
| |
Collapse
|
10
|
Payseur BA. Genetics of Recombination Rate Variation Within and Between Species. J Evol Biol 2024:voae158. [PMID: 39680417 DOI: 10.1093/jeb/voae158] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Indexed: 12/17/2024]
Abstract
Recombination diversifies the genomes of offspring, influences the evolutionary dynamics of populations, and ensures that chromosomes segregate properly during meiosis. Individuals recombine at different rates but observed levels of variation in recombination rate remain mostly unexplained. Genetic dissection of differences in recombination rate within and between species provides a powerful framework for understanding how this trait evolves. In this Perspective, I amalgamate published findings from genetic studies of variation in the genome-wide number of crossovers within and between species, and I use exploratory analyses to identify preliminary patterns. The narrow-sense heritability of crossover count is consistently low, indicating limited resemblance among relatives and predicting a weak response to short-term selection. Variants associated with crossover number within populations span the range of minor allele frequency. The size of the additive effect of recombination-associated variants, along with a negative correlation between this effect and minor allele frequency, raises the prospect that mutations inducing phenotypic shifts larger than a few crossovers are deleterious, though the contributions of methodological biases to these patterns deserve investigation. Quantitative trait loci that contribute to differences between populations or species alter crossover number in both directions, a pattern inconsistent with selection toward a constant optimum for this trait. Building on this characterization of genetic variation in crossover number within and between species, I describe fruitful avenues for future research. Better integrating recombination rate into quantitative genetics will reveal the balance of evolutionary forces responsible for genetic variation in this trait that shapes inheritance.
Collapse
Affiliation(s)
- Bret A Payseur
- Laboratory of Genetics, University of Wisconsin-Madison, Madison, WI
| |
Collapse
|
11
|
Yang Q, Carioscia SA, Isada M, McCoy RC. Approximate Bayesian computation supports a high incidence of chromosomal mosaicism in blastocyst-stage human embryos. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.26.625484. [PMID: 39677623 PMCID: PMC11642757 DOI: 10.1101/2024.11.26.625484] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2024]
Abstract
Chromosome mis-segregation is common in human meiosis and mitosis, and the resulting aneuploidies are the leading cause of pregnancy loss. Preimplantation genetic testing for aneuploidy (PGT-A) seeks to prioritize chromosomally normal embryos for transfer based on genetic analysis of a biopsy of approximately five trophectoderm cells from blastocyst-stage in vitro fertilized (IVF) embryos. While modern PGT-A platforms classify these biopsies as aneuploid, euploid, or mosaic (possessing a mixture of normal and aneuploid cells), the underlying incidences of aneuploid, euploid, and mosaic embryos and the rates of meiotic and mitotic error that produced them remain largely unknown. To address this knowledge gap, we paired a recent method for embryo simulation with approximate Bayesian computation (ABC) to infer rates of meiotic and mitotic error that best explain published PGT-A data. By simulating from these posterior distributions, we also evaluated the chromosomal status of entire embryos. For a published clinical sample, we estimated a 39-43% probability of meiotic error per meiosis, as well as a 1.0-3.0% probability of mitotic error per mitosis, depending on assumptions about spatial clustering of aneuploid cells within mosaic embryos. In addition, our analyses suggest that less than 1% of blastocysts are fully euploid, and that many embryos possess low-level mosaic clones that are not captured during biopsy. These broad conclusions were relatively insensitive to potential misclassification of mosaic biopsies. Together, our work helps overcome the limitations of embryo biopsies to estimate the fundamental rates of cell division errors that are the main causes of human pregnancy loss.
Collapse
Affiliation(s)
- Qingya Yang
- Department of Biology, Johns Hopkins University, Baltimore, MD, USA 21218
| | - Sara A. Carioscia
- Department of Biology, Johns Hopkins University, Baltimore, MD, USA 21218
| | - Matthew Isada
- Department of Biology, Johns Hopkins University, Baltimore, MD, USA 21218
| | - Rajiv C. McCoy
- Department of Biology, Johns Hopkins University, Baltimore, MD, USA 21218
| |
Collapse
|
12
|
Wang C, Chen Z, Copenhaver GP, Wang Y. Heterochromatin in plant meiosis. Nucleus 2024; 15:2328719. [PMID: 38488152 PMCID: PMC10950279 DOI: 10.1080/19491034.2024.2328719] [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: 09/11/2023] [Accepted: 03/05/2024] [Indexed: 03/19/2024] Open
Abstract
Heterochromatin is an organizational property of eukaryotic chromosomes, characterized by extensive DNA and histone modifications, that is associated with the silencing of transposable elements and repetitive sequences. Maintaining heterochromatin is crucial for ensuring genomic integrity and stability during the cell cycle. During meiosis, heterochromatin is important for homologous chromosome synapsis, recombination, and segregation, but our understanding of meiotic heterochromatin formation and condensation is limited. In this review, we focus on the dynamics and features of heterochromatin and how it condenses during meiosis in plants. We also discuss how meiotic heterochromatin influences the interaction and recombination of homologous chromosomes during prophase I.
Collapse
Affiliation(s)
- Cong Wang
- Guangdong Provincial Key Laboratory of Protein Function and Regulation in Agricultural Organisms, College of Life Sciences, South China Agricultural University, Guangzhou, China
| | - Zhiyu Chen
- State Key Laboratory of Genetic Engineering, Institute of Plant Biology, School of Life Sciences, Fudan University, Shanghai, China
| | - Gregory P. Copenhaver
- Department of Biology and the Integrative Program for Biological and Genome Sciences, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Lineberger Comprehensive Cancer Center, University of North Carolina School of Medicine, Chapel Hill, NC, USA
| | - Yingxiang Wang
- Guangdong Provincial Key Laboratory of Protein Function and Regulation in Agricultural Organisms, College of Life Sciences, South China Agricultural University, Guangzhou, China
- State Key Laboratory of Genetic Engineering, Institute of Plant Biology, School of Life Sciences, Fudan University, Shanghai, China
- Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou, China
| |
Collapse
|
13
|
Ochando I, Urbano A, Rueda J. Genetics in Reproductive Medicine. Arch Med Res 2024; 55:103092. [PMID: 39342776 DOI: 10.1016/j.arcmed.2024.103092] [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: 07/02/2024] [Revised: 09/08/2024] [Accepted: 09/18/2024] [Indexed: 10/01/2024]
Abstract
Thanks to advances in technology, genetic testing is now available to explore the causes of infertility and to assess the risk of a given couple passing on a genetic disorder to their offspring. This allows at-risk couples to make an informed decision when opting for assisted reproduction and allows professionals to offer pre-implantation diagnosis when appropriate. Genetic screening of an infertile couple has thus become standard practice for an appropriate diagnosis, treatment, and prognostic assessment. This review aims to highlight the conditions under which genetic screening plays a role in improving reproductive outcomes for infertile couples.
Collapse
Affiliation(s)
- Isabel Ochando
- Nuuma Genetics, Hospital HLA Vistahermosa, Alicante, Spain; Departamento de Histología y Anatomía, Universidad Miguel Hernández, Alicante, Spain.
| | - Antonio Urbano
- Nuuma Genetics, Hospital HLA Vistahermosa, Alicante, Spain; Departamento de Histología y Anatomía, Universidad Miguel Hernández, Alicante, Spain
| | - Joaquín Rueda
- Departamento de Histología y Anatomía, Universidad Miguel Hernández, Alicante, Spain
| |
Collapse
|
14
|
Delaval A, Glover KA, Solberg MF, Fjelldal PG, Hansen T, Harvey AC. Chromosomal aberrations and early mortality in a non-mammalian vertebrate: example from pressure-induced triploid Atlantic salmon. Heredity (Edinb) 2024; 133:426-436. [PMID: 39369146 PMCID: PMC11589116 DOI: 10.1038/s41437-024-00727-9] [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: 03/28/2024] [Revised: 09/20/2024] [Accepted: 09/23/2024] [Indexed: 10/07/2024] Open
Abstract
In commercial aquaculture, the production of triploid fish is currently the most practical approach to prevent maturation and farm-to-wild introgression following escapes. However, triploids often exhibit poor welfare, and the underlying mechanisms remain unclear. Inheritance issues associated with sub-optimal hydrostatic pressure treatments used to induce triploidy, or the genetic background of parental fish, have been speculated to contribute. We tested this by quantifying the frequency and type of chromosomal aberrations in Atlantic salmon subjected to a gradient of sub-optimal pressure treatments (Experiment 1) and from multiple mothers (Experiment 2). From these experiments, we genotyped a subsample of ~900 eyed eggs and all ~3300 surviving parr across ~20 microsatellites. In contrast to the low frequency of chromosomal aberrations in the diploid (no hydrostatic pressure) and triploid (full 9500 PSI treatment) controls, eyed eggs subjected to sub-optimal pressure treatments (6500-8500 PSI) had a higher incidence of chromosomal aberrations such as aneuploidy and uniparental disomy, corresponding to lower triploidization success and higher egg mortality rates. We also observed maternal effects on triploidization success and incidence of chromosomal aberrations, with certain half-sibling families exhibiting more aberrations than others. Chromosomal aberrations were rare among surviving parr, suggesting a purge of maladapted individuals during early development. This study demonstrates that sub-optimal hydrostatic pressure treatments and maternal effects not only influence the success of triploidization treatments, but may also affect the incidence of chromosomal aberrations and early mortality. The results have important implications for aquaculture breeding programs and their efforts to prevent farm-to-wild introgression.
Collapse
Affiliation(s)
| | | | | | | | - Tom Hansen
- Institute of Marine Research, Matre Research Station, Matredal, Norway
| | | |
Collapse
|
15
|
Fan LH, Qi ST, Wang ZB, Ouyang YC, Lei WL, Wang Y, Li A, Wang F, Li J, Li L, Li YY, Hou Y, Schatten H, Wang WH, Sun QY, Ou XH. MEIKIN expression and its C-terminal phosphorylation by PLK1 is closely related the metaphase-anaphase transition by affecting cyclin B1 and Securin stabilization in meiotic oocyte. Histochem Cell Biol 2024; 162:447-464. [PMID: 39093409 DOI: 10.1007/s00418-024-02316-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/22/2024] [Indexed: 08/04/2024]
Abstract
Oocyte meiotic maturation failure and chromosome abnormality is one of the main causes of infertility, abortion, and diseases. The mono-orientation of sister chromatids during the first meiosis is important for ensuring accurate chromosome segregation in oocytes. MEIKIN is a germ cell-specific protein that can regulate the mono-orientation of sister chromatids and the protection of the centromeric cohesin complex during meiosis I. Here we found that MEIKIN is a maternal protein that was highly expressed in mouse oocytes before the metaphase I (MI) stage, but became degraded by the MII stage and dramatically reduced after fertilization. Strikingly, MEIKIN underwent phosphorylation modification after germinal vesicle breakdown (GVBD), indicating its possible function in subsequent cellular event regulation. We further showed that MEIKIN phosphorylation was mediated by PLK1 at its carboxyl terminal region and its C-terminus was its key functional domain. To clarify the biological significance of meikin degradation during later stages of oocyte maturation, exogenous expression of MEIKIN was employed, which showed that suppression of MEIKIN degradation resulted in chromosome misalignment, cyclin B1 and Securin degradation failure, and MI arrest through a spindle assembly checkpoint (SAC)-independent mechanism. Exogenous expression of MEIKIN also inhibited metaphase II (MII) exit and early embryo development. These results indicate that proper MEIKIN expression level and its C-terminal phosphorylation by PLK1 are critical for regulating the metaphase-anaphase transition in meiotic oocyte. The findings of this study are important for understanding the regulation of chromosome segregation and the prevention meiotic abnormality.
Collapse
Affiliation(s)
- Li-Hua Fan
- Fertility Preservation Laboratory, Reproductive Medicine Center, Guangdong Second Provincial General Hospital, Guangzhou, 510317, China
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
- Guangdong-Hong Kong Metabolism & Reproduction Joint Laboratory, Guangdong Second Provincial General Hospital, Guangzhou, 510317, China
- University of Chinese Academy of Sciences, Beijing, 100101, China
| | - Shu-Tao Qi
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
- Key Laboratory of Major Obstetrics Diseases of Guangdong Province, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150, China
| | - Zhen-Bo Wang
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
- University of Chinese Academy of Sciences, Beijing, 100101, China
| | - Ying-Chun Ouyang
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Wen-Long Lei
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
- University of Chinese Academy of Sciences, Beijing, 100101, China
| | - Yue Wang
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
- University of Chinese Academy of Sciences, Beijing, 100101, China
| | - Ang Li
- Fertility Preservation Laboratory, Reproductive Medicine Center, Guangdong Second Provincial General Hospital, Guangzhou, 510317, China
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
- Guangdong-Hong Kong Metabolism & Reproduction Joint Laboratory, Guangdong Second Provincial General Hospital, Guangzhou, 510317, China
| | - Feng Wang
- Fertility Preservation Laboratory, Reproductive Medicine Center, Guangdong Second Provincial General Hospital, Guangzhou, 510317, China
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
- Guangdong-Hong Kong Metabolism & Reproduction Joint Laboratory, Guangdong Second Provincial General Hospital, Guangzhou, 510317, China
| | - Jian Li
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Li Li
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
- University of Chinese Academy of Sciences, Beijing, 100101, China
| | - Yuan-Yuan Li
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Yi Hou
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Heide Schatten
- Department of Veterinary Pathobiology, University of Missouri, Columbia, MO, 65211, USA
| | - Wei-Hua Wang
- Key Laboratory of Major Obstetrics Diseases of Guangdong Province, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150, China.
| | - Qing-Yuan Sun
- Fertility Preservation Laboratory, Reproductive Medicine Center, Guangdong Second Provincial General Hospital, Guangzhou, 510317, China.
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China.
- Guangdong-Hong Kong Metabolism & Reproduction Joint Laboratory, Guangdong Second Provincial General Hospital, Guangzhou, 510317, China.
- University of Chinese Academy of Sciences, Beijing, 100101, China.
| | - Xiang-Hong Ou
- Fertility Preservation Laboratory, Reproductive Medicine Center, Guangdong Second Provincial General Hospital, Guangzhou, 510317, China.
- Guangdong-Hong Kong Metabolism & Reproduction Joint Laboratory, Guangdong Second Provincial General Hospital, Guangzhou, 510317, China.
| |
Collapse
|
16
|
Klemm C, Ólafsson G, Wood HR, Mellor C, Zabet NR, Thorpe PH. Proteome-wide forced interactions reveal a functional map of cell-cycle phospho-regulation in S. cerevisiae. Nucleus 2024; 15:2420129. [PMID: 39618027 PMCID: PMC11622623 DOI: 10.1080/19491034.2024.2420129] [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: 07/25/2024] [Revised: 10/16/2024] [Accepted: 10/17/2024] [Indexed: 12/08/2024] Open
Abstract
Dynamic protein phosphorylation and dephosphorylation play an essential role in cell cycle progression. Kinases and phosphatases are generally highly conserved across eukaryotes, underlining their importance for post-translational regulation of substrate proteins. In recent years, advances in phospho-proteomics have shed light on protein phosphorylation dynamics throughout the cell cycle, and ongoing progress in bioinformatics has significantly improved annotation of specific phosphorylation events to a given kinase. However, the functional impact of individual phosphorylation events on cell cycle progression is often unclear. To address this question, we used the Synthetic Physical Interactions (SPI) method, which enables the systematic recruitment of phospho-regulators to most yeast proteins. Using this method, we identified several putative novel targets involved in chromosome segregation and cytokinesis. The SPI method monitors cell growth and, therefore, serves as a tool to determine the impact of protein phosphorylation on cell cycle progression.
Collapse
Affiliation(s)
- Cinzia Klemm
- School of Biological and Behavioural Sciences, Queen Mary University of London, London, UK
- Department of Bioengineering, Imperial College London, London, UK
| | - Guðjón Ólafsson
- School of Biological and Behavioural Sciences, Queen Mary University of London, London, UK
- Department of Biochemistry and Molecular Biology, BioMedical Center, Faculty of Medicine, University of Iceland, Reykjavík, Iceland
| | - Henry Richard Wood
- School of Biological and Behavioural Sciences, Queen Mary University of London, London, UK
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Caitlin Mellor
- School of Biological and Behavioural Sciences, Queen Mary University of London, London, UK
| | - Nicolae Radu Zabet
- Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Peter Harold Thorpe
- School of Biological and Behavioural Sciences, Queen Mary University of London, London, UK
| |
Collapse
|
17
|
Leem J, Gowett M, Bolarinwa S, Mogessie B. On the origin of mitosis-derived human embryo aneuploidy. Nat Commun 2024; 15:10391. [PMID: 39613785 DOI: 10.1038/s41467-024-54953-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Accepted: 11/26/2024] [Indexed: 12/01/2024] Open
Affiliation(s)
- Jiyeon Leem
- Department of Molecular, Cellular, and Developmental Biology, Yale University, New Haven, CT, USA
| | - Madison Gowett
- Department of Molecular, Cellular, and Developmental Biology, Yale University, New Haven, CT, USA
| | - Sarah Bolarinwa
- Department of Molecular, Cellular, and Developmental Biology, Yale University, New Haven, CT, USA
| | - Binyam Mogessie
- Department of Molecular, Cellular, and Developmental Biology, Yale University, New Haven, CT, USA.
| |
Collapse
|
18
|
Sharma N, Coticchio G, Borini A, Tachibana K, Nasmyth KA, Schuh M. Changes in DNA repair compartments and cohesin loss promote DNA damage accumulation in aged oocytes. Curr Biol 2024; 34:5131-5148.e6. [PMID: 39437784 DOI: 10.1016/j.cub.2024.09.040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Revised: 07/20/2024] [Accepted: 09/16/2024] [Indexed: 10/25/2024]
Abstract
Oocyte loss, a natural process that accelerates as women approach their mid-30s, poses a significant challenge to female reproduction. Recent studies have identified DNA damage as a primary contributor to oocyte loss, but the mechanisms underlying DNA damage accumulation remain unclear. Here, we show that aged oocytes have a lower DNA repair capacity and reduced mobility of DNA damage sites compared to young oocytes. Incomplete DNA repair in aged oocytes results in defective chromosome integrity and partitioning, thereby compromising oocyte quality. We found that DNA repair proteins are arranged in spatially distinct DNA repair compartments that form during the late stages of oocyte growth, accompanied by changes in the activity of DNA repair pathways. We demonstrate alterations in these compartments with age, including substantial changes in the levels of key DNA repair proteins and a shift toward error-prone DNA repair pathways. In addition, we show that reduced cohesin levels make aged oocytes more vulnerable to persistent DNA damage and cause changes in DNA repair compartments. Our study links DNA damage accumulation in aged oocytes, a leading cause of oocyte loss, to cohesin deterioration and changes in the organization, abundance, and response of DNA repair machinery.
Collapse
Affiliation(s)
- Ninadini Sharma
- Max Planck Institute for Multidisciplinary Sciences, Am Fassberg 11, Göttingen 37077, Germany
| | | | - Andrea Borini
- IVIRMA Global Research Alliance, 9.baby, Bologna 40125, Italy
| | - Kikuë Tachibana
- Department of Totipotency, Max Planck Institute of Biochemistry, Am Klopferspitz 18, Martinsried, Munich 82152, Germany
| | - Kim A Nasmyth
- Department of Biochemistry, University of Oxford, Oxford OX1 3QU, UK
| | - Melina Schuh
- Max Planck Institute for Multidisciplinary Sciences, Am Fassberg 11, Göttingen 37077, Germany.
| |
Collapse
|
19
|
Zheng Z, Jiang W, Geng T, Yang Y, Zhao C, Ling X. Diminished ovarian reserve may have no effect on embryo euploidy: A retrospective cohort study. Int J Gynaecol Obstet 2024. [PMID: 39548794 DOI: 10.1002/ijgo.16033] [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: 08/27/2024] [Revised: 10/23/2024] [Accepted: 10/29/2024] [Indexed: 11/18/2024]
Abstract
OBJECTIVE Does an association exist between diminished ovarian reserve (DOR) and embryonic aneuploidy risk in young women of reproductive age? METHODS This retrospective cohort study included 854 participants treated in the Department of Reproductive Medicine, Women's Hospital of Nanjing Medicine University, Nanjing Maternity and Child Health Care Hospital, from May 2016 to September 2023 for a total of 894 cycles. The relationship between ovarian reserve and embryonic euploidy was evaluated using statistical methods such as generalized estimating equation (GEE) and propensity score matching (PSM). RESULTS Despite the higher number of biopsied blastocysts in women with normal ovarian reserve, no significant difference in embryonic aneuploidy rates was observed between DOR women and the control group. In addition, there were no significant disparities observed in the rates of metaphase II (MII) oocytes, normal fertilization, transferable embryos, blastocyst formation, and high-scoring blastocysts between the DOR and control groups after PSM. CONCLUSION This study showed that DOR had no significant effect on the incidence of aneuploidy in embryos. Young women informed about DOR should be relatively confident about the quality of their eggs and the euploidy status of their embryos before conventional assisted reproduction, thereby mitigating concern regarding adverse maternal outcomes due to aneuploidy.
Collapse
Affiliation(s)
- Zichen Zheng
- Department of Reproductive Medicine, Women's Hospital of Nanjing Medical University, Nanjing Women and Children's Healthcare Hospital, Nanjing, China
| | - Wei Jiang
- Department of Reproductive Medicine, Women's Hospital of Nanjing Medical University, Nanjing Women and Children's Healthcare Hospital, Nanjing, China
| | - Tangyi Geng
- Department of Reproductive Medicine, Women's Hospital of Nanjing Medical University, Nanjing Women and Children's Healthcare Hospital, Nanjing, China
| | - Ye Yang
- Department of Reproductive Medicine, Women's Hospital of Nanjing Medical University, Nanjing Women and Children's Healthcare Hospital, Nanjing, China
| | - Chun Zhao
- Department of Reproductive Medicine, Women's Hospital of Nanjing Medical University, Nanjing Women and Children's Healthcare Hospital, Nanjing, China
| | - Xiufeng Ling
- Department of Reproductive Medicine, Women's Hospital of Nanjing Medical University, Nanjing Women and Children's Healthcare Hospital, Nanjing, China
| |
Collapse
|
20
|
Christensen P, Fischer R, Schulze W, Baukloh V, Kienast K, Coull G, Parner ET. Role of intra-individual variation in the detection of thresholds for DFI and for misclassification rates: A retrospective analysis of 14,775 SCSA ® tests. Andrology 2024. [PMID: 39545580 DOI: 10.1111/andr.13801] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Revised: 10/31/2024] [Accepted: 11/03/2024] [Indexed: 11/17/2024]
Abstract
BACKGROUND Sperm DNA damage is associated with reduced male fertility after natural conception and intrauterine insemination. However, the impact on in vitro fertilization (IVF) and especially intracytoplasmic sperm injection (ICSI) treatments is still unclear. Few studies have focused on the intra-individual variation in DFI even though it may have an important role to play in terms of detection of thresholds and for misclassification rates. METHODS Results for Sperm Chromatin Structure Assay (SCSA®) tests performed for 70 European fertility clinics between January 1st, 2008 and December 31st, 2022 were examined. A small retrospective study included 406 couples receiving their first treatment with IVF or ICSI. These results were then used for a mathematical simulation to investigate the role of intra-individual variation. The large retrospective study included a total of 14,138 diagnostic tests and 637 tests from an IUI study. The distribution of DFI was assessed for the IUI cohort and cohorts of patients attending Sims IVF and Fertility Center Hamburg (FCH). Descriptive analysis of the data was performed regarding time of year, male age, and year. RESULTS When DFI was above the thresholds of 15 and 25, a significant reduction in ongoing pregnancies after 12 weeks of gestation was observed for IVF and ICSI treatments, respectively. For IVF treatments, the pregnancy rate was reduced from 45.1% to 24.6%, odds ratio = 2.58 (p = 0.004). For ICSI treatments, the pregnancy rate was reduced from 48.6% to 29.6%, odds ratio = 2.00 (p = 0.032). Intra-individual variation was significantly related to the misclassification rate and the sample size required to identify a threshold. The percentage of patients with a DFI below 15 was 64.8% for the IUI cohort and 51.7% and 41.6% for cohorts of patients attending Sims IVF and FCH, respectively. The median DFI for these cohorts differed significantly and was 11.6, 15.0 and 17.2, respectively. DFI shows a seasonal variation, and increases with male age. During the past 15 years, the median DFI has increased by 0.05% per year (p = 0.02). DISCUSSION AND CONCLUSIONS Ongoing pregnancy rates are reduced significantly for both IVF and ICSI treatments when DFI is above the thresholds of 15 and 25, respectively. The misclassification rate and the required sample size increase with increasing intra-individual variation. Couples with a DFI above 15 are more likely to experience failed assisted reproductive technology (ART) cycles. DFI appears to have increased during the past 15 years.
Collapse
Affiliation(s)
| | | | | | - Vera Baukloh
- Fertility Center Hamburg, Speersort 4, Hamburg, Germany
| | | | | | - Erik T Parner
- Aarhus University, Department of Public Health, Section for Biostatistics, Bartholins Allé 2, Aarhus C, Denmark
| |
Collapse
|
21
|
Morgan AP, Payseur BA. Genetic background affects the strength of crossover interference in house mice. Genetics 2024; 228:iyae146. [PMID: 39241112 PMCID: PMC11538424 DOI: 10.1093/genetics/iyae146] [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: 07/29/2024] [Accepted: 08/22/2024] [Indexed: 09/08/2024] Open
Abstract
Meiotic recombination is required for faithful chromosome segregation in most sexually reproducing organisms and shapes the distribution of genetic variation in populations. Both the overall rate and the spatial distribution of crossovers vary within and between species. Adjacent crossovers on the same chromosome tend to be spaced more evenly than expected at random, a phenomenon known as crossover interference. Although interference has been observed in many taxa, the factors that influence the strength of interference are not well understood. We used house mice (Mus musculus), a well-established model system for understanding recombination, to study the effects of genetics and age on recombination rate and interference in the male germline. We analyzed crossover positions in 503 progeny from reciprocal F1 hybrids between inbred strains representing the three major subspecies of house mice. Consistent with previous studies, autosomal alleles from M. m. musculus tend to increase recombination rate, while inheriting a M. m. musculus X chromosome decreases recombination rate. Old males transmit an average of 0.6 more crossovers per meiosis (5.0%) than young males, though the effect varies across genetic backgrounds. We show that the strength of crossover interference depends on genotype, providing a rare demonstration that interference evolves over short timescales. Differences between reciprocal F1s suggest that X-linked factors modulate the strength of interference. Our findings motivate additional comparisons of interference among recently diverged species and further examination of the role of paternal age in determining the number and positioning of crossovers.
Collapse
Affiliation(s)
- Andrew P Morgan
- Department of Medicine, University of North Carolina, Campus Box #7085, Chapel Hill, NC 27599-7085, USA
| | - Bret A Payseur
- Laboratory of Genetics, University of Wisconsin, Madison, WI 53706, USA
| |
Collapse
|
22
|
Figliuzzi M, Bori L, Ottolini CS, Picchetta L, Caroselli S, Reverenna M, Poli M, Campbell A, Smith R, Coticchio G, Cimadomo D, Rienzi LF, Meseguer M, Capalbo A. Human embryos with segmental aneuploidies display delayed early development: a multicenter morphokinetic analysis. Fertil Steril 2024:S0015-0282(24)02344-6. [PMID: 39510239 DOI: 10.1016/j.fertnstert.2024.10.042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Revised: 10/21/2024] [Accepted: 10/28/2024] [Indexed: 11/15/2024]
Abstract
OBJECTIVE To assess whether segmental aneuploid embryos display unique morphokinetic patterns. DESIGN Retrospective multicenter study including a total of 7,027 embryos cultured between 2016 and 2021 in three European in vitro fertilization centers. Analysis was performed on aggregated multicenter data and separately for data from each center. Embryos with no more than four chromosomal alterations were considered in the analysis, resulting in 3,040 euploids and 2,818 whole-chromosome and 697 segmental aneuploids. Overall, the data set contained 3,742 distinct euploid-segmental sibling pairs. SETTING In vitro fertilization clinics. PATIENT(S) Standard morphokinetic features were annotated using various time-lapse systems. Blastocysts were subjected to comprehensive chromosomal screening via preimplantation genetic testing for aneuploidy. INTERVENTION(S) Morphokinetic patterns were compared among euploid, whole-chromosome aneuploid, and segmental aneuploid embryos. MAIN OUTCOME MEASURE(S) Morphokinetic timings across groups were compared using statistical analysis, and associations with cleavage features were assessed. Multicenter and center-specific multivariate logistic regression models were calibrated, and their predictive performance was evaluated on independent test set data using area under the receiver operating characteristic curve (AUROC) metrics. RESULT(S) Segmental aneuploid embryos cleaved significantly slower than their euploid siblings across the first three cell cycles, with a delay reaching the blastocyst-stage of development. Specifically during these early cell cycles, segmental aneuploid embryos were also shown to be significantly slower than their aneuploid siblings. A logistic model on the basis of morphokinetic data from the multicenter data set and regressed against type of aneuploidy displayed modest predictive performance on an independent test set (train-AUROC = 0.58; test-AUROC = 0.57). Predictive performance improved on the basis of data from a single center displaying adequate predictive performance on an independent test set from the same center (train-AUROC = 0.74; test-AUROC = 0.64). However, the predictive value diminished when tested on data from other centers (AUROC = 0.52-0.55). Finally, the presence of multinucleation and blastomere exclusion at the cleavage stage were associated with segmental aneuploidies. The combination of morphokinetic features and these discrete embryo morphological features into the logistic regression model (train-AUROC = 0.71) provided an improved prediction of segmental aneuploidy, supporting future investigations using more comprehensive annotation systems. CONCLUSION(S) The developed predictive framework may help improve decision-making in preimplantation genetic testing for aneuploidy cycles, helping in the evaluation of embryos showing segmental aneuploidy and distinguishing which embryos are more likely to not have lethal uniform aneuploidies for transfer.
Collapse
Affiliation(s)
| | - Lorena Bori
- IVIRMA Global Research Alliance, IVI, Valencia, Spain
| | - Christian Simon Ottolini
- Juno Genetics Italy, Reproductive Genetics, Rome, Italy; Department of Maternal and Fetal Medicine, UCL, London, United Kingdom
| | | | | | - Marco Reverenna
- Università di Pavia, Master Genomic Data Science, Pavia, Italy
| | - Maurizio Poli
- Juno Genetics Italy, Reproductive Genetics, Rome, Italy
| | | | | | | | - Danilo Cimadomo
- IVIRMA Global Research Alliance, Genera, Clinica Valle Giulia, Rome, Italy
| | - Laura Francesca Rienzi
- IVIRMA Global Research Alliance, Genera, Clinica Valle Giulia, Rome, Italy; Department of Biomolecular Sciences, University of Urbino "Carlo Bo," Urbino, Italy
| | | | - Antonio Capalbo
- Juno Genetics Italy, Reproductive Genetics, Rome, Italy; Unit of Molecular Genetics, Center for Advanced Studies and Technology (CAST), "G. d'Annunzio" University of Chieti-Pescara, Chieti, Italy; Department of Psychological Health and Territorial Sciences, School of Medicine and Health Sciences, "G. d'Annunzio" University of Chieti-Pescara, Chieti, Italy.
| |
Collapse
|
23
|
Biswas L, Tyc KM, Aboelenain M, Sun S, Dundović I, Vukušić K, Liu J, Guo V, Xu M, Scott RT, Tao X, Tolić IM, Xing J, Schindler K. Maternal genetic variants in kinesin motor domains prematurely increase egg aneuploidy. Proc Natl Acad Sci U S A 2024; 121:e2414963121. [PMID: 39475646 PMCID: PMC11551467 DOI: 10.1073/pnas.2414963121] [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: 07/26/2024] [Accepted: 09/27/2024] [Indexed: 11/06/2024] Open
Abstract
The female reproductive lifespan is highly dependent on egg quality, especially the presence of a normal number of chromosomes in an egg, known as euploidy. Mistakes in meiosis leading to egg aneuploidy are frequent in humans. Yet, knowledge of the precise genetic landscape that causes egg aneuploidy in women is limited, as phenotypic data on the frequency of human egg aneuploidy are difficult to obtain and therefore absent in public genetic datasets. Here, we identify genetic determinants of reproductive aging via egg aneuploidy in women using a biobank of individual maternal exomes linked with maternal age and embryonic aneuploidy data. Using the exome data, we identified 404 genes bearing variants enriched in individuals with pathologically elevated egg aneuploidy rates. Analysis of the gene ontology and protein-protein interaction network implicated genes encoding the kinesin protein family in egg aneuploidy. We interrogate the causal relationship of the human variants within candidate kinesin genes via experimental perturbations and demonstrate that motor domain variants increase aneuploidy in mouse oocytes. Finally, using a knock-in mouse model, we validate that a specific variant in kinesin KIF18A accelerates reproductive aging and diminishes fertility. These findings reveal additional functional mechanisms of reproductive aging and shed light on how genetic variation underlies individual heterogeneity in the female reproductive lifespan, which might be leveraged to predict reproductive longevity. Together, these results lay the groundwork for the noninvasive biomarkers for egg quality, a first step toward personalized fertility medicine.
Collapse
Affiliation(s)
- Leelabati Biswas
- Department of Genetics, Rutgers, The State University of New Jersey, Piscataway, NJ 08854
- Human Genetics Institute of New Jersey, Rutgers, The State University of New Jersey, Piscataway, NJ 08854
- Rutgers Robert Wood Johnson Medical School, Rutgers, The State University of New Jersey, Piscataway, NJ 08854
| | - Katarzyna M Tyc
- Department of Genetics, Rutgers, The State University of New Jersey, Piscataway, NJ 08854
- Human Genetics Institute of New Jersey, Rutgers, The State University of New Jersey, Piscataway, NJ 08854
| | - Mansour Aboelenain
- Department of Genetics, Rutgers, The State University of New Jersey, Piscataway, NJ 08854
- Human Genetics Institute of New Jersey, Rutgers, The State University of New Jersey, Piscataway, NJ 08854
- Department of Theriogenology, Faculty of Veterinary Medicine, Mansoura University, Mansoura 35516, Egypt
| | - Siqi Sun
- Department of Genetics, Rutgers, The State University of New Jersey, Piscataway, NJ 08854
- Human Genetics Institute of New Jersey, Rutgers, The State University of New Jersey, Piscataway, NJ 08854
| | - Iva Dundović
- Department of Molecular Biology, Ruđer Bošković Institute, Zagreb 1000, Croatia
| | - Kruno Vukušić
- Department of Molecular Biology, Ruđer Bošković Institute, Zagreb 1000, Croatia
| | - Jason Liu
- Department of Genetics, Rutgers, The State University of New Jersey, Piscataway, NJ 08854
| | | | - Min Xu
- Department of Statistics, Rutgers, The State University of New Jersey, Piscataway, NJ 08854
| | - Richard T Scott
- Foundation for Embryonic Competence, Basking Ridge, NJ 07920
| | - Xin Tao
- Juno Genetics US, Basking Ridge, NJ 07920
| | - Iva M Tolić
- Department of Molecular Biology, Ruđer Bošković Institute, Zagreb 1000, Croatia
| | - Jinchuan Xing
- Department of Genetics, Rutgers, The State University of New Jersey, Piscataway, NJ 08854
- Human Genetics Institute of New Jersey, Rutgers, The State University of New Jersey, Piscataway, NJ 08854
| | - Karen Schindler
- Department of Genetics, Rutgers, The State University of New Jersey, Piscataway, NJ 08854
- Human Genetics Institute of New Jersey, Rutgers, The State University of New Jersey, Piscataway, NJ 08854
| |
Collapse
|
24
|
Fouks Y, Vaughan D, Sripada V, Penzias AS, Bortoletto P, Sakkas D. Do sperm factors influence embryonic aneuploidy? Long live the oocyte. Hum Reprod 2024; 39:2442-2452. [PMID: 39352944 DOI: 10.1093/humrep/deae224] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Revised: 08/07/2024] [Indexed: 10/04/2024] Open
Abstract
STUDY QUESTION What is the impact of male age- and sperm-related factors on embryonic aneuploidy? SUMMARY ANSWER Using a 3-fold analysis framework encompassing patient-level, embryo-level, and matching analysis, we found no clinically significant interactions between male age and sperm quality with embryo ploidy. WHAT IS KNOWN ALREADY While the effect of maternal age on embryo chromosomal aneuploidy is well-established, the impact of male age and sperm quality on ploidy is less well-defined. STUDY DESIGN, SIZE, DURATION This retrospective cohort study analyzed autologous preimplantation genetic testing for aneuploidy (PGT-A) and frozen embryo transfer cycles from December 2014 to June 2021. The study involved 11 087 cycles from 8484 patients, with a total of 35 797 embryos. PARTICIPANTS/MATERIALS, SETTING, METHODS The aneuploidy rate, calculated as the ratio of aneuploid blastocysts to the total number of blastocysts biopsied in a single treatment cycle, was evaluated. In the embryo-level analysis, the main outcome measure was the ploidy state of the embryos. The study employed a multifaceted analytical approach that included a patient-level analysis using generalized linear mixed models, an embryo-level analysis focusing on chromosomal ploidy, and a propensity score matching analysis contrasting groups with distinct ploidy rates (0% and 100%). There were no interventions as this was an observational study of PGT-A cycles. MAIN RESULTS AND THE ROLE OF CHANCE No clinically relevant factors influencing ploidy rate related to male and sperm quality were revealed. In contrast, female age (coefficient = -0.053), BMI (coefficient = 0.003), prior ART cycle (coefficient = -0.066), and number of oocytes retrieved (coefficient = -0.018) were identified at the patient level. Embryo analysis identified age (coefficient = -0.1244) and ICSI usage (coefficient = -0.0129) as significant factors. Despite these, no significant interactions between male and female assessed factors on the ploidy rate emerged. Propensity score matching between maximal (100% vs 0%) euploid rates did not reveal significant differences of influence by male age and sperm quality. LIMITATIONS, REASONS FOR CAUTION The focus on patients having blastocyst biopsy for PGT-A may not reflect the broader IVF population. Other semen quality parameters like DNA fragmentation were not included. Exclusion of embryo mosaicism from the analysis could affect aneuploidy rate interpretations. There may also be unmeasured influences like lifestyle or environmental factors. WIDER IMPLICATIONS OF THE FINDINGS Male age and sperm quality parameters were consistent across both maximal and minimal ploidy rate comparisons. No significant clinical characteristics related to the factors assessed for the male-influenced blastocyst ploidy status, confirming the dominancy of the oocyte and female age. STUDY FUNDING/COMPETING INTEREST(S) The study was not funded. There are no competing interests. TRIAL REGISTRATION NUMBER N/A.
Collapse
Affiliation(s)
- Yuval Fouks
- Boston IVF, IVIRMA Global Research Alliance, Waltham, MA, USA
- Reproductive Services, The Royal Women's Hospital, Parkville, Victoria, Australia
- The Department of Gynecology and Obstetrics, Faculty of Medical and Health Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Denis Vaughan
- Boston IVF, IVIRMA Global Research Alliance, Waltham, MA, USA
- Department of Obstetrics and Gynecology, Beth Israel Deaconess Medical Center, Boston, MA, USA
- Department of Obstetrics, Gynecology and Reproductive Biology, Harvard Medical School, Boston, MA, USA
| | - Veda Sripada
- Harvard T.H. Chan School of Public Health, Cambridge, MA, USA
| | - Alan S Penzias
- Boston IVF, IVIRMA Global Research Alliance, Waltham, MA, USA
- Department of Obstetrics and Gynecology, Beth Israel Deaconess Medical Center, Boston, MA, USA
- Department of Obstetrics, Gynecology and Reproductive Biology, Harvard Medical School, Boston, MA, USA
| | - Pietro Bortoletto
- Boston IVF, IVIRMA Global Research Alliance, Waltham, MA, USA
- Department of Obstetrics and Gynecology, Beth Israel Deaconess Medical Center, Boston, MA, USA
- Department of Obstetrics, Gynecology and Reproductive Biology, Harvard Medical School, Boston, MA, USA
| | - Denny Sakkas
- Boston IVF, IVIRMA Global Research Alliance, Waltham, MA, USA
| |
Collapse
|
25
|
Beebeejaun Y, Nicolaides KH, Mania A, Sarris I, Sunkara SK. Preimplantation Genetic Testing for Aneuploidy (PGT-A) in In-Vitro Fertilisation (IVF) Treatment: Study Protocol for Pilot Phase of a Randomised Controlled Trial. J Clin Med 2024; 13:6192. [PMID: 39458142 PMCID: PMC11508202 DOI: 10.3390/jcm13206192] [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: 07/23/2024] [Revised: 08/30/2024] [Accepted: 10/09/2024] [Indexed: 10/28/2024] Open
Abstract
Introduction: Poor outcomes following IVF treatments are speculated to be due to the transfer of aneuploid embryos that cannot be identified based on morphological evaluation alone. This leads to patients requiring numerous embryo transfers and, consequently, a prolonged time interval before live birth. Embryo selection following preimplantation genetic testing for aneuploidy (PGT-A) with next-generation sequencing (NGS) has been suggested as an intervention to shorten time to pregnancy in women undergoing in vitro fertilisation (IVF). Past studies assessing the clinical efficacy of PGT-A in improving clinical outcomes have been conflicting and the associated clinical pregnancy rates and live birth rates following the transfer of a mosaic embryos have yet to be determined. None of the existing studies solely included women of advanced reproductive age (ARA). The pilot study and proposed RCT will determine if, compared to morphological evaluation alone, the use of PGT-A through NGS is a more clinically effective, safer, and more cost-effective way to provide IVF treatment in women of advanced reproductive age. Method and Analysis: The proposed pilot study will aim to randomise 100 patients within a single-centre study to evaluate recruitment, randomisation, and adherence to study protocol and allocated trail arms by participating patients. The results of the pilot study will enable us to determine the sample size for a larger study to establish the effectiveness of PGT-A in ARA women. Ethics and Dissemination: The study (Integrated Research Application System Number 236067) received approval from the Health Research Authority and Health and Care Research Wales (HCRW) and the East Midlands-Leicester South Research Ethics Committee (20/EM/0290). The results will be made available to patients, the funders, the Reproductive Medicine societies, and other researchers. Trial registration: ClinicalTrials.gov Identifier: NCT05009745, n.
Collapse
Affiliation(s)
- Yusuf Beebeejaun
- King’s Fertility, Fetal Medicine Research Institute, King’s College Hospital, London SE5 8BB, UK
- Department of Women’s and Children’s Health, Faculty of Life Sciences and Medicine, King’s College London, London SE1 7EH, UK
| | - Kypros H. Nicolaides
- Harris Birthright Research Centre of Fetal Medicine, Fetal Medicine Research Institute, King’s College Hospital, London SE5 8BB, UK
| | - Anastasia Mania
- King’s Fertility, Fetal Medicine Research Institute, King’s College Hospital, London SE5 8BB, UK
| | - Ippokratis Sarris
- King’s Fertility, Fetal Medicine Research Institute, King’s College Hospital, London SE5 8BB, UK
- Department of Women’s and Children’s Health, Faculty of Life Sciences and Medicine, King’s College London, London SE1 7EH, UK
| | - Sesh K. Sunkara
- King’s Fertility, Fetal Medicine Research Institute, King’s College Hospital, London SE5 8BB, UK
- Department of Women’s and Children’s Health, Faculty of Life Sciences and Medicine, King’s College London, London SE1 7EH, UK
| |
Collapse
|
26
|
Ryan CA, Purfield DC, Matthews D, Rathje C, Valldecabres A, Berry DP. Prevalence of Autosomal Monosomy and Trisomy Estimated Using Single Nucleotide Polymorphism Genotype Intensity Chip Information in a Large Population of Juvenile Dairy and Beef Cattle. J Anim Breed Genet 2024. [PMID: 39400955 DOI: 10.1111/jbg.12902] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Revised: 09/09/2024] [Accepted: 09/16/2024] [Indexed: 10/15/2024]
Abstract
Aneuploidy, a genetic condition characterised by the deletion (monosomy) or duplication (trisomy) of a chromosome, has been extensively studied in humans, particularly in the context of trisomy on chromosome 21, also known as Down syndrome. Research on autosomal aneuploidy in live-born cattle has been limited to case reports, resulting in a lack of prevalence estimates of aneuploidy in cattle. Furthermore, the viability or lethality of aneuploidy on specific autosomes in cattle has not been well documented. The objective of this study was to estimate the prevalence of autosomal aneuploidy in a large population of new-born and juvenile beef and dairy cattle using single nucleotide polymorphism (SNP) chip genotype intensity data. Of the population of 779,138 cattle genotyped when younger than 15 months of age, 139 cattle (i.e., 0.017%) were diagnosed with one case of autosomal trisomy. Trisomy in only 10 different autosomes were detected (BTA 4, 6, 12, 15, 20, 24, 26, 27, 28 and 29) albeit the one case of trisomy detected on Bos taurus autosome (BTA) 4 was in an additional population of 341,927 cattle that were genotyped at > 15 months of age and was therefore excluded from prevalence estimates to minimise bias. The prevalence of trisomy per chromosome was generally inversely related to the length of the chromosome. Although the number of affected individuals was few, there was no evidence of differences in prevalence by breed, inbreeding level or parental age. The parental origin of the detected cases of trisomy was maternal for 92% of the cases. No cases of monosomy were detected despite the large dataset, which included calves genotyped at birth, indicating the potential lethal nature of monosomy in cattle. Cytogenetic testing was used to verify three of the animals with detected autosomal trisomy who were still alive. Eighteen of the 139 animals identified with autosomal trisomy were recorded as being stillborn, resulting in a prevalence of autosomal aneuploidy in live-born cattle of 0.015%. Of the 121 live-born cattle with autosomal trisomy, a total of 68 died on farm at, on average (standard deviation), 6.8 (8.7) months of age. All animals with autosomal trisomy on BTA 6, 12, 15, 20 or 24 were either stillborn or died on farm within 15 days of birth. This study is the first report of trisomy on BTA 4, 6, 15, 20 and 27 in live-born cattle, as well as the first to document fertile cows with trisomy on BTA 4, 27 or 28. Given that genotype intensity SNP data from SNP-chips are readily available, identifying animals affected with autosomal aneuploidy as well as quantifying and monitoring the incidence can be easily undertaken.
Collapse
Affiliation(s)
- Cliona A Ryan
- Department of Animal Bioscience, Teagasc, Moorepark, Co. Cork, Ireland
- Department of Biological Science, Munster Technological University, Co. Cork, Ireland
| | - Deirdre C Purfield
- Department of Biological Science, Munster Technological University, Co. Cork, Ireland
| | - Daragh Matthews
- Genetics Department, Irish Cattle Breeding Federation, Co. Cork, Ireland
| | - Claudia Rathje
- School of Biosciences, University of Kent, Canterbury, UK
| | | | - Donagh P Berry
- Department of Animal Bioscience, Teagasc, Moorepark, Co. Cork, Ireland
| |
Collapse
|
27
|
Rojas J, Hose J, Dutcher HA, Place M, Wolters JF, Hittinger CT, Gasch AP. Comparative modeling reveals the molecular determinants of aneuploidy fitness cost in a wild yeast model. CELL GENOMICS 2024; 4:100656. [PMID: 39317188 PMCID: PMC11602619 DOI: 10.1016/j.xgen.2024.100656] [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: 04/23/2024] [Revised: 07/10/2024] [Accepted: 08/20/2024] [Indexed: 09/26/2024]
Abstract
Although implicated as deleterious in many organisms, aneuploidy can underlie rapid phenotypic evolution. However, aneuploidy will be maintained only if the benefit outweighs the cost, which remains incompletely understood. To quantify this cost and the molecular determinants behind it, we generated a panel of chromosome duplications in Saccharomyces cerevisiae and applied comparative modeling and molecular validation to understand aneuploidy toxicity. We show that 74%-94% of the variance in aneuploid strains' growth rates is explained by the cumulative cost of genes on each chromosome, measured for single-gene duplications using a genomic library, along with the deleterious contribution of small nucleolar RNAs (snoRNAs) and beneficial effects of tRNAs. Machine learning to identify properties of detrimental gene duplicates provided no support for the balance hypothesis of aneuploidy toxicity and instead identified gene length as the best predictor of toxicity. Our results present a generalized framework for the cost of aneuploidy with implications for disease biology and evolution.
Collapse
Affiliation(s)
- Julie Rojas
- Center for Genomic Science Innovation, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - James Hose
- Center for Genomic Science Innovation, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - H Auguste Dutcher
- Center for Genomic Science Innovation, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Michael Place
- Center for Genomic Science Innovation, University of Wisconsin-Madison, Madison, WI 53706, USA; Great Lakes Bioenergy Research Center, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - John F Wolters
- Laboratory of Genetics, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Chris Todd Hittinger
- Center for Genomic Science Innovation, University of Wisconsin-Madison, Madison, WI 53706, USA; Great Lakes Bioenergy Research Center, University of Wisconsin-Madison, Madison, WI 53706, USA; Laboratory of Genetics, University of Wisconsin-Madison, Madison, WI 53706, USA; J.F. Crow Institute for the Study of Evolution, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Audrey P Gasch
- Center for Genomic Science Innovation, University of Wisconsin-Madison, Madison, WI 53706, USA; Great Lakes Bioenergy Research Center, University of Wisconsin-Madison, Madison, WI 53706, USA; Laboratory of Genetics, University of Wisconsin-Madison, Madison, WI 53706, USA; J.F. Crow Institute for the Study of Evolution, University of Wisconsin-Madison, Madison, WI 53706, USA.
| |
Collapse
|
28
|
Wang J, So C. Aged eggs improve within young follicles. NATURE AGING 2024; 4:1338-1339. [PMID: 39354178 DOI: 10.1038/s43587-024-00715-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/03/2024]
Affiliation(s)
- Jing Wang
- National Institute of Biological Sciences, Beijing (NIBS), Beijing, China
- Tsinghua Institute of Multidisciplinary Biomedical Research, Tsinghua University, Beijing, China
| | - Chun So
- National Institute of Biological Sciences, Beijing (NIBS), Beijing, China.
- Tsinghua Institute of Multidisciplinary Biomedical Research, Tsinghua University, Beijing, China.
- Department of Obstetrics and Gynecology, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong.
| |
Collapse
|
29
|
Cozzolino M, Capalbo A, Garcia-Velasco JA, Pellicer A, Vaiarelli A, Galliano D, Cimadomo D, Ubaldi FM, Parini V, Levi-Setti PE. In vitro fertilization and perinatal outcomes of patients with advanced maternal age after single frozen euploid embryo transfer: a propensity score-matched analysis of autologous and donor cycles. Fertil Steril 2024; 122:678-686. [PMID: 38838805 DOI: 10.1016/j.fertnstert.2024.05.170] [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: 09/30/2023] [Revised: 05/29/2024] [Accepted: 05/30/2024] [Indexed: 06/07/2024]
Abstract
OBJECTIVE To evaluate in vitro fertilization (IVF) and perinatal outcomes of donor egg and autologous cycles in patients with advanced reproductive age after undergoing single frozen euploid embryo transfer. DESIGN A multicenter, retrospective, cohort study. SETTING University-affiliated and private IVF centers. PATIENT(S) Patients aged 39-46 years who underwent IVF with intracytoplasmic sperm injection and preimplantation genetic testing for aneuploidy using whole-chromosome sequencing with donor (n = 278) or autologous (n = 278) oocytes between October 2017 and October 2021. INTERVENTION(S) Single frozen euploid embryo transfer with donor or autologous euploid embryo. MAIN OUTCOME MEASURE(S) The main outcome measure was the live birth rate (LBR) after the first embryo transfer, calculated per embryo transfer. The secondary outcomes included the implantation rate, ectopic pregnancy rate, miscarriage rate, and gestational age and birth weight at the time of delivery. RESULT(S) Patients using donor or autologous oocytes had a similar likelihood of implantation (57.91% [51.87-63.78] vs. 57.19% [51.15-63.09]) and LBR (41.01% [95% confidence interval {CI}, 35.17-47.04] vs. 42.45% [95% CI, 36.56-48.49]). Furthermore, there were no significant differences in the ectopic pregnancy rate (0.72% [0.09-2.57] vs. 0.36% [0.01-1.99]), miscarriage rate (16.19% [12.06-21.05] vs. 14.39% [95% CI, 10.48-19.08]), gestational age (38.50 [38.08-38.92] vs. 39.16 [38.25-40.07] weeks), or birth weight of infants (2,982.25 [2,606.69-3,357.81] vs. 3,128.24 [2,962.30-3,294.17] kg). The univariate analysis showed no association between advanced maternal age and the LBR (relative risk, 1.03 [95% CI, 0.84-1.25]). Multivariate analysis using putative confounders for embryo competency found no associations with LBR (adjusted relative risk, 1.22 [95% CI, 0.75-1.98]). CONCLUSION(S) Patients with euploid blastocysts derived from donor or autologous oocytes did not reveal statistically significant differences in the LBR, implantation rate, ectopic pregnancy rate, miscarriage rate, duration of gestation, or infant birth weight. These findings suggest that age-related reproductive decline and/or poor IVF outcomes associated with women with advanced reproductive age undergoing IVF are heavily driven by embryonic aneuploidy.
Collapse
Affiliation(s)
- Mauro Cozzolino
- IVIRMA Global Research Alliance, IVI Roma, Rome, Italy; IVIRMA Global Research Alliance, IVI Foundation, Instituto de Investigación Sanitaria La Fe (IIS La Fe), Valencia, Spain.
| | - Antonio Capalbo
- Juno Genetics, Rome, Italy; Department of Psychological Health and Territorial Sciences, School of Medicine and Health Sciences, "G. d'Annunzio" University of Chieti-Pescara, Chieti, Italy; Unit of Medical Genetics, Center for Advanced Studies and Technology (CAST), "G. d'Annunzio" University of Chieti-Pescara, Chieti, Italy
| | | | - Antonio Pellicer
- IVIRMA Global Research Alliance, IVI Roma, Rome, Italy; IVIRMA Global Research Alliance, IVI Foundation, Instituto de Investigación Sanitaria La Fe (IIS La Fe), Valencia, Spain
| | - Alberto Vaiarelli
- IVIRMA Global Research Alliance, Genera, Clinica Valle Giulia, Rome, Italy
| | | | - Danilo Cimadomo
- IVIRMA Global Research Alliance, Genera, Clinica Valle Giulia, Rome, Italy
| | | | | | - Paolo Emanuele Levi-Setti
- IRCCS Humanitas Research Hospital - Rozzano, Milan, Italy; Department of Biomedical Sciences, Humanitas University, Milan, Italy
| |
Collapse
|
30
|
Staniczek J, Manasar-Dyrbuś M, Rybak-Krzyszkowska M, Kondracka A, Orszulak D, Niziński K, Sadłocha M, Drosdzol-Cop A, Stojko R, Illa M. Systematic review and meta-analysis of the association between young maternal age and fetal abnormalities. Sci Rep 2024; 14:22562. [PMID: 39343828 PMCID: PMC11439902 DOI: 10.1038/s41598-024-74015-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Accepted: 09/23/2024] [Indexed: 10/01/2024] Open
Abstract
Fetal anomalies among young women and adolescents pose major public health concerns. This systematic review aims to elucidate the relationship between young maternal age and the incidence of fetal abnormalities. According to the systematic review and meta-analysis PRISMA protocol, cohort, cross-sectional and case-control studies were scrutinized to include 80,393,450 participants across diverse regions. The meta-analysis utilized Odds Ratios (OR) as the effect measure, adopting a random-effects model. The screening process involved 157 selected and verified manuscripts, which ultimately resulted in the final inclusion of 20 studied in the meta-analysis. The criterion for young maternal age was the age of ≤ 20 years. The meta-analysis revealed a pooled OR of 0.93 (95% CI: 0.82-1.05, p = 0.252), indicating no statistically significant association between young maternal age (≤ 20 years) and fetal anomalies. However, considerable heterogeneity (I² = 96.21%) was noted, prompting the use of a random-effects model to derive the reported results. The meta-analysis did not find statistically significant differences in the occurrence of congenital anomalies in fetuses of younger women than in overall population. Although due to significant heterogeneity of the analyzed studies, and a risk of bias, caution should be exercised when interpreting the results, further investigation may be warranted to understand the relationship between maternal age and risk of fetal anomalies. Nevertheless, the study shows significant differences, which diminish in collective analysis, suggests that factors beyond age may be influential. Specifically, the limited access to or quality of healthcare in certain regions could be a more critical factor than maternal age itself.
Collapse
Affiliation(s)
- Jakub Staniczek
- Department of Gynecology, Obstetrics and Gynecological Oncology, Medical University of Silesia, Katowice, 40-211, Poland.
| | - Maisa Manasar-Dyrbuś
- Department of Gynecology, Obstetrics and Gynecological Oncology, Medical University of Silesia, Katowice, 40-211, Poland
| | | | - Adrianna Kondracka
- Department of Obstetrics and Pathology of Pregnancy, Medical University of Lublin, Lublin, 20-059, Poland
| | - Dominika Orszulak
- Department of Gynecology, Obstetrics and Gynecological Oncology, Medical University of Silesia, Katowice, 40-211, Poland
| | - Kacper Niziński
- Department of Gynecology, Obstetrics and Gynecological Oncology, Medical University of Silesia, Katowice, 40-211, Poland
| | - Marcin Sadłocha
- Department of Gynecology, Obstetrics and Gynecological Oncology, Medical University of Silesia, Katowice, 40-211, Poland
| | - Agnieszka Drosdzol-Cop
- Department of Gynecology, Obstetrics and Gynecological Oncology, Medical University of Silesia, Katowice, 40-211, Poland
| | - Rafał Stojko
- Department of Gynecology, Obstetrics and Gynecological Oncology, Medical University of Silesia, Katowice, 40-211, Poland
| | - Miriam Illa
- BCNatal | Fetal Medicine Research Center (Hospital Clínic and Hospital Sant Joan de Déu), Universitat de Barcelona, Barcelona, 08028, Spain
| |
Collapse
|
31
|
Cornelisse S, Fleischer K, van der Westerlaken L, de Bruin JP, Vergouw C, Koks C, Derhaag J, Visser J, van Echten-Arends J, Slappendel E, Arends B, van der Zanden M, van Dongen A, Brink-van der Vlugt J, de Hundt M, Curfs M, Verhoeve H, Traas-Hofmans M, Wurth Y, Manger P, Pieterse Q, Braat D, van Wely M, Ramos L, Mastenbroek S. Cumulative live birth rate of a blastocyst versus cleavage stage embryo transfer policy during in vitro fertilisation in women with a good prognosis: multicentre randomised controlled trial. BMJ 2024; 386:e080133. [PMID: 39284610 PMCID: PMC11403767 DOI: 10.1136/bmj-2024-080133] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 07/24/2024] [Indexed: 09/22/2024]
Abstract
OBJECTIVES To evaluate whether embryo transfers at blastocyst stage improve the cumulative live birth rate after oocyte retrieval, including both fresh and frozen-thawed transfers, and whether the risk of obstetric and perinatal complications is increased compared with cleavage stage embryo transfers during in vitro fertilisation (IVF) treatment. DESIGN Multicentre randomised controlled trial. SETTING 21 hospitals and clinics in the Netherlands, 18 August 2018 to 17 December 2021. PARTICIPANTS 1202 women with at least four embryos available on day 2 after oocyte retrieval were randomly assigned to either blastocyst stage embryo transfer (n=603) or cleavage stage embryo transfer (n=599). INTERVENTIONS In the blastocyst group and cleavage group, embryo transfers were performed on day 5 and day 3, respectively, after oocyte retrieval, followed by cryopreservation of surplus embryos. Analysis was on an intention-to-treat basis, with secondary analyses as per protocol. MAIN OUTCOME MEASURES The primary outcome was the cumulative live birth rate per oocyte retrieval, including results of all frozen-thawed embryo transfers within a year after randomisation. Secondary outcomes included cumulative rates of pregnancy, pregnancy loss, and live birth after fresh embryo transfer, number of embryo transfers needed, number of frozen embryos, and obstetric and perinatal outcomes. RESULTS The cumulative live birth rate did not differ between the blastocyst group and cleavage group (58.9% (355 of 603) v 58.4% (350 of 599; risk ratio 1.01, 95% confidence interval (CI) 0.84 to 1.22). The blastocyst group showed a higher live birth rate after fresh embryo transfer (1.26, 1.00 to 1.58), lower cumulative pregnancy loss rate (0.68, 0.51 to 0.89), and lower mean number of embryo transfers needed to result in a live birth (1.55 v 1.82; P<0.001). The incidence of moderate preterm birth (32 to <37 weeks) in singletons was higher in the blastocyst group (1.87, 1.05 to 3.34). CONCLUSION Blastocyst stage embryo transfers resulted in a similar cumulative live birth rate to cleavage stage embryo transfers in women with at least four embryos available during IVF treatment. TRIAL REGISTRATION International Clinical Trial Registry Platform NTR7034.
Collapse
Affiliation(s)
- Simone Cornelisse
- Department of Obstetrics and Gynaecology, Radboud University Medical Centre, Nijmegen, Netherlands
| | | | | | - Jan-Peter de Bruin
- Department of Obstetrics and Gynaecology, Jeroen Bosch Ziekenhuis, 's-Hertogenbosch, Netherlands
| | - Carlijn Vergouw
- Amsterdam UMC location, Vrije Universiteit Amsterdam, Centre for Reproductive Medicine Amsterdam, Netherlands
| | - Carolien Koks
- Department of Obstetrics and Gynaecology, Maxima Medical Centre, Veldhoven, Netherlands
| | - Josien Derhaag
- Centre for Reproductive Medicine, MUMC+, Maastricht, Netherlands
| | - Jantien Visser
- Department of Obstetrics and Gynaecology, Amphia Ziekenhuis, Breda, Netherlands
| | | | - Els Slappendel
- Centre for Fertility, Nij Geertgen, Elsendorp, Netherlands
| | - Brigitte Arends
- Department of Reproductive Medicine, University Medical Centre Utrecht, Utrecht, Netherlands
| | - Moniek van der Zanden
- Department of Obstetrics and Gynaecology, Haaglanden Medical Centre, The Hague, Netherlands
| | - Angelique van Dongen
- Department of Obstetrics and Gynaecology, Hospital Gelderse Vallei, Ede, Netherlands
| | | | - Marcella de Hundt
- Department of Obstetrics and Gynaecology, NoordWest Ziekenhuisgroep, Alkmaar, Netherlands
| | - Max Curfs
- Isala Fertility Centre, Isala Clinics, Zwolle, Netherlands
| | - Harold Verhoeve
- Department of Obstetrics and Gynaecology, OLVG Oost, Amsterdam, Netherlands
| | - Maaike Traas-Hofmans
- Department of Obstetrics and Gynaecology, Gelre Ziekenhuizen, Apeldoorn and Zutphen, Netherlands
| | - Yvonne Wurth
- IVF Centre, Elisabeth-TweeSteden Ziekenhuis, Tilburg, Netherlands
| | - Petra Manger
- Department of Obstetrics and Gynaecology, Diakonessenhuis, Utrecht, Netherlands
| | - Quirine Pieterse
- Department of Obstetrics and Gynaecology, Haga Ziekenhuis, Den Haag, Netherlands
| | - Didi Braat
- Department of Obstetrics and Gynaecology, Radboud University Medical Centre, Nijmegen, Netherlands
| | - Madelon van Wely
- Amsterdam UMC, location University of Amsterdam, Centre for Reproductive Medicine, Amsterdam, Netherlands
- Amsterdam Reproduction and Development Research Institute, Amsterdam, Netherlands
| | - Liliana Ramos
- Department of Obstetrics and Gynaecology, Radboud University Medical Centre, Nijmegen, Netherlands
| | - Sebastiaan Mastenbroek
- Amsterdam UMC, location University of Amsterdam, Centre for Reproductive Medicine, Amsterdam, Netherlands
- Amsterdam Reproduction and Development Research Institute, Amsterdam, Netherlands
| |
Collapse
|
32
|
Chen X, Zhang X, Jiang T, Xu W. Klinefelter syndrome: etiology and clinical considerations in male infertility†. Biol Reprod 2024; 111:516-528. [PMID: 38785325 DOI: 10.1093/biolre/ioae076] [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] [Revised: 05/08/2024] [Indexed: 05/25/2024] Open
Abstract
Klinefelter syndrome (KS) is the most prevalent chromosomal disorder occurring in males. It is defined by an additional X chromosome, 47,XXY, resulting from errors in chromosomal segregation during parental gametogenesis. A major phenotype is impaired reproductive function, in the form of low testosterone and infertility. This review comprehensively examines the genetic and physiological factors contributing to infertility in KS, in addition to emergent assisted reproductive technologies, and the unique ethical challenges KS patients face when seeking infertility treatment. The pathology underlying KS is increased susceptibility for meiotic errors during spermatogenesis, resulting in aneuploid or even polyploid gametes. Specific genetic elements potentiating this susceptibility include polymorphisms in checkpoint genes regulating chromosomal synapsis and segregation. Physiologically, the additional sex chromosome also alters testicular endocrinology and metabolism by dysregulating interstitial and Sertoli cell function, collectively impairing normal sperm development. Additionally, epigenetic modifications like aberrant DNA methylation are being increasingly implicated in these disruptions. We also discuss assisted reproductive approaches leveraged in infertility management for KS patients. Application of assisted reproductive approaches, along with deep comprehension of the meiotic and endocrine disturbances precipitated by supernumerary X chromosomes, shows promise in enabling biological parenthood for KS individuals. This will require continued multidisciplinary collaboration between experts with background of genetics, physiology, ethics, and clinical reproductive medicine.
Collapse
Affiliation(s)
- Xinyue Chen
- Reproductive Endocrinology and Regulation Laboratory, Department of Obstetric and Gynecologic, West China Second University Hospital, Sichuan University, Chengdu 610041, China
| | - Xueguang Zhang
- Reproductive Endocrinology and Regulation Laboratory, Department of Obstetric and Gynecologic, West China Second University Hospital, Sichuan University, Chengdu 610041, China
| | - Ting Jiang
- Reproductive Endocrinology and Regulation Laboratory, Department of Obstetric and Gynecologic, West China Second University Hospital, Sichuan University, Chengdu 610041, China
| | - Wenming Xu
- Reproductive Endocrinology and Regulation Laboratory, Department of Obstetric and Gynecologic, West China Second University Hospital, Sichuan University, Chengdu 610041, China
- Key Laboratory of Obstetric, Gynecologic and Pediatric Diseases and Birth Defects of Ministry of Education, West China Second University Hospital, Sichuan University-The Chinese University of Hong Kong (SCU-CUHK) Joint Laboratory for Reproductive Medicine, Chengdu 610041, China
| |
Collapse
|
33
|
Deryabin PI, Borodkina AV. The Role of the Endometrium in Implantation: A Modern View. Int J Mol Sci 2024; 25:9746. [PMID: 39273693 PMCID: PMC11395593 DOI: 10.3390/ijms25179746] [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: 08/02/2024] [Revised: 09/06/2024] [Accepted: 09/08/2024] [Indexed: 09/15/2024] Open
Abstract
According to the current data, the endometrium acts as a "sensor" of embryo quality, which promotes the implantation of euploid embryos and prevents the implantation and/or subsequent development of genetically abnormal embryos. The present review addresses the nature of the "sensory function" of the endometrium and highlights the necessity for assessing its functional status. The first section examines the evolutionary origin of the "sensory" ability of the endometrium as a consequence of spontaneous decidualization that occurred in placental animals. The second section details the mechanisms for implementing this function at the cellular level. In particular, the recent findings of the appearance of different cell subpopulations during decidualization are described, and their role in implantation is discussed. The pathological consequences of an imbalance among these subpopulations are also discussed. Finally, the third section summarizes information on currently available clinical tools to assess endometrial functional status. The advantages and disadvantages of the approaches are emphasized, and possible options for developing more advanced technologies for assessing the "sensory" function of the endometrium are proposed.
Collapse
Affiliation(s)
- Pavel I Deryabin
- Mechanisms of Cellular Senescence Laboratory, Institute of Cytology of the Russian Academy of Sciences, Tikhoretsky Ave. 4, Saint-Petersburg 194064, Russia
| | - Aleksandra V Borodkina
- Mechanisms of Cellular Senescence Laboratory, Institute of Cytology of the Russian Academy of Sciences, Tikhoretsky Ave. 4, Saint-Petersburg 194064, Russia
| |
Collapse
|
34
|
Weng Q, Wan L, Straker GC, Deegan TD, Duncker BP, Neiman AM, Luk E, Hollingsworth NM. An acidic loop in the forkhead-associated domain of the yeast meiosis-specific kinase Mek1 interacts with a specific motif in a subset of Mek1 substrates. Genetics 2024; 228:iyae106. [PMID: 38979911 PMCID: PMC11373509 DOI: 10.1093/genetics/iyae106] [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: 05/24/2024] [Revised: 06/27/2024] [Accepted: 06/28/2024] [Indexed: 07/10/2024] Open
Abstract
The meiosis-specific kinase Mek1 regulates key steps in meiotic recombination in the budding yeast, Saccharomyces cerevisiae. MEK1 limits resection at double-strand break (DSB) ends and is required for preferential strand invasion into homologs, a process known as interhomolog bias. After strand invasion, MEK1 promotes phosphorylation of the synaptonemal complex protein Zip1 that is necessary for DSB repair mediated by a crossover-specific pathway that enables chromosome synapsis. In addition, Mek1 phosphorylation of the meiosis-specific transcription factor, Ndt80, regulates the meiotic recombination checkpoint that prevents exit from pachytene when DSBs are present. Mek1 interacts with Ndt80 through a 5-amino acid sequence, RPSKR, located between the DNA-binding and activation domains of Ndt80. AlphaFold Multimer modeling of a fragment of Ndt80 containing the RPSKR motif and full-length Mek1 indicated that RPSKR binds to an acidic loop located in the Mek1 FHA domain, a noncanonical interaction with this motif. A second protein, the 5'-3' helicase Rrm3, similarly interacts with Mek1 through an RPAKR motif and is an in vitro substrate of Mek1. Genetic analysis using various mutants in the MEK1 acidic loop validated the AlphaFold model, in that they specifically disrupt 2-hybrid interactions with Ndt80 and Rrm3. Phenotypic analyses further showed that the acidic loop mutants are defective in the meiotic recombination checkpoint and, in certain circumstances, exhibit more severe phenotypes compared to the NDT80 mutant with the RPSKR sequence deleted, suggesting that additional, as yet unknown, substrates of Mek1 also bind to Mek1 using an RPXKR motif.
Collapse
Affiliation(s)
- Qixuan Weng
- Department of Biochemistry and Cell Biology, Stony Brook University, Stony Brook, NY 11794-5215, USA
| | - Lihong Wan
- Department of Biochemistry and Cell Biology, Stony Brook University, Stony Brook, NY 11794-5215, USA
| | - Geburah C Straker
- Department of Biology, University of Waterloo, Waterloo, Ontario N2L 3G1, Canada
| | - Tom D Deegan
- MRC Protein Phosphorylation and Ubiquitylation Unit, Sir James Black Centre, School of Life Sciences, University of Dundee, Dundee DD1 5EH, UK
- MRC Human Genetics Unit, Institute of Genetics and Cancer, University of Edinburgh, Western General Hospital, Edinburgh EH4 2XU, UK
| | - Bernard P Duncker
- Department of Biology, University of Waterloo, Waterloo, Ontario N2L 3G1, Canada
| | - Aaron M Neiman
- Department of Biochemistry and Cell Biology, Stony Brook University, Stony Brook, NY 11794-5215, USA
| | - Ed Luk
- Department of Biochemistry and Cell Biology, Stony Brook University, Stony Brook, NY 11794-5215, USA
| | - Nancy M Hollingsworth
- Department of Biochemistry and Cell Biology, Stony Brook University, Stony Brook, NY 11794-5215, USA
| |
Collapse
|
35
|
McAuley JB, Servin B, Burnett HA, Brekke C, Peters L, Hagen IJ, Niskanen AK, Ringsby TH, Husby A, Jensen H, Johnston SE. The Genetic Architecture of Recombination Rates is Polygenic and Differs Between the Sexes in Wild House Sparrows (Passer domesticus). Mol Biol Evol 2024; 41:msae179. [PMID: 39183719 PMCID: PMC11385585 DOI: 10.1093/molbev/msae179] [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: 03/20/2024] [Revised: 06/01/2024] [Accepted: 07/11/2024] [Indexed: 08/27/2024] Open
Abstract
Meiotic recombination through chromosomal crossing-over is a fundamental feature of sex and an important driver of genomic diversity. It ensures proper disjunction, allows increased selection responses, and prevents mutation accumulation; however, it is also mutagenic and can break up favorable haplotypes. This cost-benefit dynamic is likely to vary depending on mechanistic and evolutionary contexts, and indeed, recombination rates show huge variation in nature. Identifying the genetic architecture of this variation is key to understanding its causes and consequences. Here, we investigate individual recombination rate variation in wild house sparrows (Passer domesticus). We integrate genomic and pedigree data to identify autosomal crossover counts (ACCs) and intrachromosomal allelic shuffling (r¯intra) in 13,056 gametes transmitted from 2,653 individuals to their offspring. Females had 1.37 times higher ACC, and 1.55 times higher r¯intra than males. ACC and r¯intra were heritable in females and males (ACC h2 = 0.23 and 0.11; r¯intra h2 = 0.12 and 0.14), but cross-sex additive genetic correlations were low (rA = 0.29 and 0.32 for ACC and r¯intra). Conditional bivariate analyses showed that all measures remained heritable after accounting for genetic values in the opposite sex, indicating that sex-specific ACC and r¯intra can evolve somewhat independently. Genome-wide models showed that ACC and r¯intra are polygenic and driven by many small-effect loci, many of which are likely to act in trans as global recombination modifiers. Our findings show that recombination rates of females and males can have different evolutionary potential in wild birds, providing a compelling mechanism for the evolution of sexual dimorphism in recombination.
Collapse
Affiliation(s)
- John B McAuley
- Institute of Ecology and Evolution, School of Biological Sciences, University of Edinburgh, Edinburgh EH9 3FL, UK
| | - Bertrand Servin
- Génétique Physiologie et Systèmes d'Elevage (GenPhySE), Université de Toulouse, INRAE, ENVT, Castanet Tolosan 31326, France
| | - Hamish A Burnett
- Centre for Biodiversity Dynamics, Department of Biology, Norwegian University of Science and Technology, Trondheim 7491, Norway
| | - Cathrine Brekke
- Institute of Ecology and Evolution, School of Biological Sciences, University of Edinburgh, Edinburgh EH9 3FL, UK
| | - Lucy Peters
- Génétique Physiologie et Systèmes d'Elevage (GenPhySE), Université de Toulouse, INRAE, ENVT, Castanet Tolosan 31326, France
| | - Ingerid J Hagen
- Centre for Biodiversity Dynamics, Department of Biology, Norwegian University of Science and Technology, Trondheim 7491, Norway
- Norwegian Institute for Nature Research, Trondheim 7034, Norway
| | - Alina K Niskanen
- Centre for Biodiversity Dynamics, Department of Biology, Norwegian University of Science and Technology, Trondheim 7491, Norway
- Ecology and Genetics Research Unit, University of Oulu, Oulu 90014, Finland
| | - Thor Harald Ringsby
- Centre for Biodiversity Dynamics, Department of Biology, Norwegian University of Science and Technology, Trondheim 7491, Norway
| | - Arild Husby
- Evolutionary Biology, Department of Ecology and Genetics, Uppsala University, Uppsala 75236, Sweden
| | - Henrik Jensen
- Centre for Biodiversity Dynamics, Department of Biology, Norwegian University of Science and Technology, Trondheim 7491, Norway
| | - Susan E Johnston
- Institute of Ecology and Evolution, School of Biological Sciences, University of Edinburgh, Edinburgh EH9 3FL, UK
| |
Collapse
|
36
|
Ryan CA, Purfield DC, Matthews D, Canedo-Ribeiro C, Valldecabres A, Berry DP. Prevalence of sex-chromosome aneuploidy estimated using SNP genotype intensity information in a large population of juvenile dairy and beef cattle. J Anim Breed Genet 2024; 141:571-585. [PMID: 38551070 DOI: 10.1111/jbg.12866] [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: 11/22/2023] [Revised: 02/06/2024] [Accepted: 03/12/2024] [Indexed: 08/12/2024]
Abstract
Aneuploidy is a genetic condition characterized by the loss or gain of one or more chromosomes. Aneuploidy affecting the sex chromosomes can lead to infertility in otherwise externally phenotypically normal cattle. Early identification of cattle with sex chromosomal aneuploidy is important to minimize the costs associated with rearing infertile cattle and futile breeding attempts. As most livestock breeding programs routinely genotype their breeding populations using single nucleotide polymorphism (SNP) arrays, this study aimed to assess the feasibility of integrating an aneuploidy screening tool into the existing pipelines that handle dense SNP genotype data. A further objective was to estimate the prevalence of sex chromosome aneuploidy in a population of 146,431 juvenile cattle using available genotype intensity data. Three genotype intensity statistics were used: the LogR Ratio (LRR), R-value (the sum of X and Y SNP probe intensities), and B-allele frequency (BAF) measurements. Within the female-verified population of 124,958 individuals, the estimated prevalence rate was 0.0048% for XO, 0.0350% for XXX, and 0.0004% for XXY. The prevalence of XXY in the male-verified population was 0.0870% (i.e., 18 out of 20,670 males). Cytogenetic testing was used to verify 2 of the XXX females who were still alive. The proposed approach can be readily integrated into existing genomic pipelines, serving as an efficient, large-scale screening tool for aneuploidy. Its implementation could enable the early identification of infertile animals with sex-chromosome aneuploidy.
Collapse
Affiliation(s)
- Cliona A Ryan
- Teagasc, Moorepark, Fermoy, County Cork, Ireland
- Munster Technological University, Cork, County Cork, Ireland
| | | | - Daragh Matthews
- Irish Cattle Breeding Federation, Ballincollig, County Cork, Ireland
| | | | | | | |
Collapse
|
37
|
Kumar R, Oke A, Rockmill B, de Cruz M, Verduzco R, Shodhan A, Woodruff-Madeira X, Abrahamsson DP, Varshavsky J, Lam J, Robinson JF, Allard P, Woodruff TJ, Fung JC. Rapid identification of reproductive toxicants among environmental chemicals using an in vivo evaluation of gametogenesis in budding yeast Saccharomyces cerevisiae. Reprod Toxicol 2024; 128:108630. [PMID: 38906490 PMCID: PMC11648579 DOI: 10.1016/j.reprotox.2024.108630] [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: 03/29/2024] [Revised: 05/26/2024] [Accepted: 05/31/2024] [Indexed: 06/23/2024]
Abstract
Infertility affects ∼12 % of couples, with environmental chemical exposure as a potential contributor. Of the chemicals that are actively manufactured, very few are assessed for reproductive health effects. Rodents are commonly used to evaluate reproductive effects, which is both costly and time consuming. Thus, there is a pressing need for rapid methods to test a broader range of chemicals. Here, we developed a strategy to evaluate large numbers of chemicals for reproductive toxicity via a yeast, S. cerevisiae high-throughput assay to assess gametogenesis as a potential new approach method (NAM). By simultaneously assessing chemicals for growth effects, we can distinguish if a chemical affects gametogenesis only, proliferative growth only or both. We identified a well-known mammalian reproductive toxicant, bisphenol A (BPA) and ranked 19 BPA analogs for reproductive harm. By testing mixtures of BPA and its analogs, we found that BPE and 17 β-estradiol each together with BPA showed synergistic effects that worsened reproductive outcome. We examined an additional 179 environmental chemicals including phthalates, pesticides, quaternary ammonium compounds and per- and polyfluoroalkyl substances and found 57 with reproductive effects. Many of the chemicals were found to be strong reproductive toxicants that have yet to be tested in mammals. Chemicals having affect before meiosis I division vs. meiosis II division were identified for 16 gametogenesis-specific chemicals. Finally, we demonstrate that in general yeast reproductive toxicity correlates well with published reproductive toxicity in mammals illustrating the promise of this NAM to quickly assess chemicals to prioritize the evaluation for human reproductive harm.
Collapse
Affiliation(s)
- Ravinder Kumar
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of California, San Francisco, CA, USA; Center of Reproductive Sciences, University of California, San Francisco, CA, USA
| | - Ashwini Oke
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of California, San Francisco, CA, USA; Center of Reproductive Sciences, University of California, San Francisco, CA, USA
| | - Beth Rockmill
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of California, San Francisco, CA, USA; Center of Reproductive Sciences, University of California, San Francisco, CA, USA
| | - Matthew de Cruz
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of California, San Francisco, CA, USA; Center of Reproductive Sciences, University of California, San Francisco, CA, USA
| | - Rafael Verduzco
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of California, San Francisco, CA, USA; Center of Reproductive Sciences, University of California, San Francisco, CA, USA
| | - Anura Shodhan
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of California, San Francisco, CA, USA; Center of Reproductive Sciences, University of California, San Francisco, CA, USA
| | - Xavier Woodruff-Madeira
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of California, San Francisco, CA, USA; Center of Reproductive Sciences, University of California, San Francisco, CA, USA
| | - Dimitri P Abrahamsson
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of California, San Francisco, CA, USA
| | - Julia Varshavsky
- Department of Health Sciences and Department of Civil and Environmental Engineering, Northeastern University, Boston, MA, USA
| | - Juleen Lam
- Department of Public Health, California State University, East Bay, Hayward, CA, USA
| | - Joshua F Robinson
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of California, San Francisco, CA, USA; Center of Reproductive Sciences, University of California, San Francisco, CA, USA
| | - Patrick Allard
- UCLA Institute for Society & Genetics, University of California, Los Angeles, CA, USA; Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA, USA
| | - Tracey J Woodruff
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of California, San Francisco, CA, USA
| | - Jennifer C Fung
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of California, San Francisco, CA, USA; Center of Reproductive Sciences, University of California, San Francisco, CA, USA.
| |
Collapse
|
38
|
Kozlov G, Franceschi C, Vedunova M. Intricacies of aging and Down syndrome. Neurosci Biobehav Rev 2024; 164:105794. [PMID: 38971514 DOI: 10.1016/j.neubiorev.2024.105794] [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: 04/11/2024] [Revised: 06/26/2024] [Accepted: 07/01/2024] [Indexed: 07/08/2024]
Abstract
Down syndrome is the most frequently occurring genetic condition, with a substantial escalation in risk associated with advanced maternal age. The syndrome is characterized by a diverse range of phenotypes, affecting to some extent all levels of organization, and its progeroid nature - early manifestation of aspects of the senile phenotype. Despite extensive investigations, many aspects and mechanisms of the disease remain unexplored. The current review aims to provide an overview of the main causes and manifestations of Down syndrome, while also examining the phenomenon of accelerated aging and exploring potential therapeutic strategies.
Collapse
Affiliation(s)
- G Kozlov
- Institute of Biology and Biomedicine, Lobachevsky State University of Nizhny Novgorod, Nizhny Novgorod, Gagarin ave., 23, 603022, Russia
| | - C Franceschi
- Lobachevsky State University of Nizhny Novgorod, Nizhny Novgorod, Gagarin ave., 23, 603022, Russia
| | - M Vedunova
- Institute of Biology and Biomedicine, Lobachevsky State University of Nizhny Novgorod, Nizhny Novgorod, Gagarin ave., 23, 603022, Russia; Prokhorov General Physics Institute of the Russian Academy of Sciences, 38 Vavilov str., 119991 Moscow, Russia.
| |
Collapse
|
39
|
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
|
40
|
Kalinderi K, Kalinderis M, Papaliagkas V, Fidani L. The Reproductive Lifespan of Ovarian Follicle. Reprod Sci 2024; 31:2604-2614. [PMID: 38816594 DOI: 10.1007/s43032-024-01606-8] [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: 12/18/2023] [Accepted: 05/27/2024] [Indexed: 06/01/2024]
Abstract
The functional unit within mammalian ovaries is the ovarian follicle. The development of the ovarian follicle is a lengthy process beginning from the time of embryogenesis, passing through multiple different stages of maturation. The purpose of this review is to describe the most basic events in the journey of ovarian follicle development, discussing the importance of ovarian reserve and highlighting the role of several factors that affect oocyte quality and quantity during aging including hormonal, genetic and epigenetic factors. Novel, promising anti-aging strategies are also discussed.
Collapse
Affiliation(s)
- Kallirhoe Kalinderi
- Laboratory of Medical Biology-Genetics, School of Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki, Thessaloniki, GR-54124, Greece.
| | - Michail Kalinderis
- Department of Obstetrics and Gynaecology, St George's University Hospital NHS Trust, Blackshaw Road, Tooting, London, SW17 0QT, UK
| | - Vasileios Papaliagkas
- Department of Biomedical Sciences, School of Health Sciences, International Hellenic University, Thessaloniki, 57400, Greece
| | - Liana Fidani
- Laboratory of Medical Biology-Genetics, School of Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki, Thessaloniki, GR-54124, Greece
| |
Collapse
|
41
|
Caroselli S, Poli M, Gatta V, Stuppia L, Capalbo A. Preconception carrier screening and preimplantation genetic testing in the infertility management. Andrology 2024. [PMID: 39166614 DOI: 10.1111/andr.13744] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Revised: 07/19/2024] [Accepted: 08/10/2024] [Indexed: 08/23/2024]
Abstract
BACKGROUND Genetic testing serves as a valuable element of reproductive care, applicable at various stages of the reproductive journey: (i) before pregnancy, when a couple's genetic reproductive risk can be evaluated; (ii) before embryo implantation, as part of in vitro fertilization (IVF) treatment, to ascertain several inherited or de novo genetic/chromosomal diseases of the embryo before transfer; (iii) during the prenatal period, to assess the genetic costitution of the fetus. Preconception carrier screening (CS) is a genetic test typically performed on couples planning a pregnancy. The primary purpose of CS is to identify couples at-risk of conceiving a child affected by a severe genetic disorder with autosomal recessive or X-linked inheritance. Detection of high reproductive risk through CS allows prospective parents to be informed of their predisposition and improve reproductive decision-making. These include undergoing IVF with preimplantation genetic testing (PGT) or donor gametes, prenatal diagnosis, adoption, remaining childless, taking no actions. Both the presence of the affected gene (PGT-M) and chromosomal status (PGT-A) of the embryo can be comprehensively assessed through modern approaches. OBJECTIVES We provide a review of CS and PGT applications to equip healthcare providers with up-to-date information regarding their opportunities and complexities. RESULTS AND DISCUSSION The use of CS and PGT is currently considered the most effective intervention for avoiding both an affected pregnancy whilst using autologous gametes in couples with known increased risk, and chromosomal abnormalities. As our understanding in the genetic component in pathological conditions increases, the number of tested disorders will expand, offering a more thorough assessment of one's genetic inheritance. Nevertheless, implementation and development in this field must be accompanied by scientific and ethical considerations to ensure this approach serves the best long-term interests of individuals and society, promoting justice and autonomy and preserving parenthood and the healthcare system. CONCLUSION The combination of CS and PGT aligns with principles of personalized medicine by offering reproductive care tailored to the individual's genetic makeup.
Collapse
Affiliation(s)
- Silvia Caroselli
- Juno Genetics, Rome, Italy
- Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy
| | | | - Valentina Gatta
- Unit of Molecular Genetics, Center for Advanced Studies and Technology (CAST), "G. d'Annunzio" University of Chieti-Pescara, Chieti-Pescara, Italy
- Department of Psychological Health and Territorial Sciences, School of Medicine and Health Sciences, "G. d'Annunzio" University of Chieti-Pescara, Chieti-Pescara, Italy
| | - Liborio Stuppia
- Unit of Molecular Genetics, Center for Advanced Studies and Technology (CAST), "G. d'Annunzio" University of Chieti-Pescara, Chieti-Pescara, Italy
- Department of Psychological Health and Territorial Sciences, School of Medicine and Health Sciences, "G. d'Annunzio" University of Chieti-Pescara, Chieti-Pescara, Italy
| | - Antonio Capalbo
- Juno Genetics, Rome, Italy
- Unit of Molecular Genetics, Center for Advanced Studies and Technology (CAST), "G. d'Annunzio" University of Chieti-Pescara, Chieti-Pescara, Italy
- Department of Psychological Health and Territorial Sciences, School of Medicine and Health Sciences, "G. d'Annunzio" University of Chieti-Pescara, Chieti-Pescara, Italy
| |
Collapse
|
42
|
Chen C, Huang Z, Dong S, Ding M, Li J, Wang M, Zeng X, Zhang X, Sun X. Calcium signaling in oocyte quality and functionality and its application. Front Endocrinol (Lausanne) 2024; 15:1411000. [PMID: 39220364 PMCID: PMC11361953 DOI: 10.3389/fendo.2024.1411000] [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: 04/02/2024] [Accepted: 07/29/2024] [Indexed: 09/04/2024] Open
Abstract
Calcium (Ca2+) is a second messenger for many signal pathways, and changes in intracellular Ca2+ concentration ([Ca2+]i) are an important signaling mechanism in the oocyte maturation, activation, fertilization, function regulation of granulosa and cumulus cells and offspring development. Ca2+ oscillations occur during oocyte maturation and fertilization, which are maintained by Ca2+ stores and extracellular Ca2+ ([Ca2+]e). Abnormalities in Ca2+ signaling can affect the release of the first polar body, the first meiotic division, and chromosome and spindle morphology. Well-studied aspects of Ca2+ signaling in the oocyte are oocyte activation and fertilization. Oocyte activation, driven by sperm-specific phospholipase PLCζ, is initiated by concerted intracellular patterns of Ca2+ release, termed Ca2+ oscillations. Ca2+ oscillations persist for a long time during fertilization and are coordinately engaged by a variety of Ca2+ channels, pumps, regulatory proteins and their partners. Calcium signaling also regulates granulosa and cumulus cells' function, which further affects oocyte maturation and fertilization outcome. Clinically, there are several physical and chemical options for treating fertilization failure through oocyte activation. Additionally, various exogenous compounds or drugs can cause ovarian dysfunction and female infertility by inducing abnormal Ca2+ signaling or Ca2+ dyshomeostasis in oocytes and granulosa cells. Therefore, the reproductive health risks caused by adverse stresses should arouse our attention. This review will systematically summarize the latest research progress on the aforementioned aspects and propose further research directions on calcium signaling in female reproduction.
Collapse
Affiliation(s)
- Chen Chen
- Institute of Reproductive Medicine, Medical School, Nantong University, Nantong, China
| | - Zefan Huang
- Institute of Reproductive Medicine, Medical School, Nantong University, Nantong, China
| | - Shijue Dong
- Institute of Reproductive Medicine, Medical School, Nantong University, Nantong, China
| | - Mengqian Ding
- Institute of Reproductive Medicine, Medical School, Nantong University, Nantong, China
| | - Jinran Li
- Center for Reproductive Medicine, Affiliated Hospital of Nantong University, Nantong University, Nantong, China
| | - Miaomiao Wang
- Institute of Reproductive Medicine, Medical School, Nantong University, Nantong, China
| | - Xuhui Zeng
- Institute of Reproductive Medicine, Medical School, Nantong University, Nantong, China
| | - Xiaoning Zhang
- Institute of Reproductive Medicine, Medical School, Nantong University, Nantong, China
| | - Xiaoli Sun
- Center for Reproductive Medicine, Affiliated Hospital of Nantong University, Nantong University, Nantong, China
| |
Collapse
|
43
|
Morgan AP, Payseur BA. Genetic background affects the strength of crossover interference in house mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.28.596233. [PMID: 38854148 PMCID: PMC11160618 DOI: 10.1101/2024.05.28.596233] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2024]
Abstract
Meiotic recombination is required for faithful chromosome segregation in most sexually reproducing organisms and shapes the distribution of genetic variation in populations. Both the overall rate and the spatial distribution of crossovers vary within and between species. Adjacent crossovers on the same chromosome tend to be spaced more evenly than expected at random, a phenomenon known as crossover interference. Although interference has been observed in many taxa, the factors that influence the strength of interference are not well understood. We used house mice (Mus musculus), a well-established model system for understanding recombination, to study the effects of genetics and age on recombination rate and interference in the male germline. We analyzed crossover positions in 503 progeny from reciprocal F1 hybrids between inbred strains representing the three major subspecies of house mice. Consistent with previous studies, autosomal alleles from M. m. musculus tend to increase recombination rate, while inheriting a M. m. musculus X chromosome decreases recombination rate. Old males transmit an average of 0.6 more crossovers per meiosis (5.0%) than young males, though the effect varies across genetic backgrounds. We show that the strength of crossover interference depends on genotype, providing a rare demonstration that interference evolves over short timescales. Differences between reciprocal F1s suggest that X-linked factors modulate the strength of interference. Our findings motivate additional comparisons of interference among recently diverged species and further examination of the role of paternal age in determining the number and positioning of crossovers.
Collapse
Affiliation(s)
- Andrew P Morgan
- Department of Medicine, University of North Carolina, Chapel Hill, NC
| | - Bret A Payseur
- Laboratory of Genetics, University of Wisconsin, Madison, WI
| |
Collapse
|
44
|
Chavez-Badiola A, Farías AFS, Mendizabal-Ruiz G, Silvestri G, Griffin DK, Valencia-Murillo R, Drakeley AJ, Cohen J. Use of artificial intelligence embryo selection based on static images to predict first-trimester pregnancy loss. Reprod Biomed Online 2024; 49:103934. [PMID: 38824762 DOI: 10.1016/j.rbmo.2024.103934] [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: 10/18/2023] [Revised: 02/16/2024] [Accepted: 02/27/2024] [Indexed: 06/04/2024]
Abstract
RESEARCH QUESTION Can an artificial intelligence embryo selection assistant predict the incidence of first-trimester spontaneous abortion using static images of IVF embryos? DESIGN In a blind, retrospective study, a cohort of 172 blastocysts from IVF cases with single embryo transfer and a positive biochemical pregnancy test was ranked retrospectively by the artificial intelligence morphometric algorithm ERICA. Making use of static embryo images from a light microscope, each blastocyst was assigned to one of four possible groups (optimal, good, fair or poor), and linear regression was used to correlate the results with the presence or absence of a normal fetal heart beat as an indicator of ongoing pregnancy or spontaneous abortion, respectively. Additional analyses included modelling for recipient age and chromosomal status established by preimplantation genetic testing for aneuploidy (PGT-A). RESULTS Embryos classified as optimal/good had a lower incidence of spontaneous abortion (16.1%) compared with embryos classified as fair/poor (25%; OR = 0.46, P = 0.005). The incidence of spontaneous abortion in chromosomally normal embryos (determined by PGT-A) was 13.3% for optimal/good embryos and 20.0% for fair/poor embryos, although the difference was not significant (P = 0.531). There was a significant association between embryo rank and recipient age (P = 0.018), in that the incidence of spontaneous abortion was unexpectedly lower in older recipients (21.3% for age ≤35 years, 17.9% for age 36-38 years, 16.4% for age ≥39 years; OR = 0.354, P = 0.0181). Overall, these results support correlation between risk of spontaneous abortion and embryo rank as determined by artificial intelligence; classification accuracy was calculated to be 67.4%. CONCLUSIONS This preliminary study suggests that artificial intelligence (ERICA), which was designed as a ranking system to assist with embryo transfer decisions and ploidy prediction, may also be useful to provide information for couples on the risk of spontaneous abortion. Future work will include a larger sample size and karyotyping of miscarried pregnancy tissue.
Collapse
Affiliation(s)
- Alejandro Chavez-Badiola
- University of Kent, School of Biosciences, Canterbury, UK; IVF 2.0 Ltd, London, UK; New Hope Fertility Center, Guadalajara, Mexico; Conceivable Life Sciences, New York, NY, USA
| | | | - Gerardo Mendizabal-Ruiz
- Conceivable Life Sciences, New York, NY, USA; Departamento de Ciencias Computacionales, Universidad de Guadalajara, Guadalajara, Mexico
| | - Giuseppe Silvestri
- University of Kent, School of Biosciences, Canterbury, UK; Conceivable Life Sciences, New York, NY, USA
| | | | | | - Andrew J Drakeley
- IVF 2.0 Ltd, London, UK; Hewitt Fertility Centre, Liverpool Women's NHS Foundation Trust, Liverpool, UK
| | - Jacques Cohen
- IVF 2.0 Ltd, London, UK; Conceivable Life Sciences, New York, NY, USA
| |
Collapse
|
45
|
Pannafino G, Chen JJ, Mithani V, Payero L, Gioia M, Crickard JB, Alani E. The Dmc1 recombinase physically interacts with and promotes the meiotic crossover functions of the Mlh1-Mlh3 endonuclease. Genetics 2024; 227:iyae066. [PMID: 38657110 PMCID: PMC11228845 DOI: 10.1093/genetics/iyae066] [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: 11/12/2023] [Revised: 04/16/2024] [Accepted: 04/17/2024] [Indexed: 04/26/2024] Open
Abstract
The accurate segregation of homologous chromosomes during the Meiosis I reductional division in most sexually reproducing eukaryotes requires crossing over between homologs. In baker's yeast approximately 80% of meiotic crossovers result from Mlh1-Mlh3 and Exo1 acting to resolve double-Holliday junction intermediates in a biased manner. Little is known about how Mlh1-Mlh3 is recruited to recombination intermediates to perform its role in crossover resolution. We performed a gene dosage screen in baker's yeast to identify novel genetic interactors with Mlh1-Mlh3. Specifically, we looked for genes whose lowered dosage reduced meiotic crossing over using sensitized mlh3 alleles that disrupt the stability of the Mlh1-Mlh3 complex and confer defects in mismatch repair but do not disrupt meiotic crossing over. To our surprise we identified genetic interactions between MLH3 and DMC1, the recombinase responsible for recombination between homologous chromosomes during meiosis. We then showed that Mlh3 physically interacts with Dmc1 in vitro and in vivo. Partial complementation of Mlh3 crossover functions was observed when MLH3 was expressed under the control of the CLB1 promoter (NDT80 regulon), suggesting that Mlh3 function can be provided late in meiotic prophase at some functional cost. A model for how Dmc1 could facilitate Mlh1-Mlh3's role in crossover resolution is presented.
Collapse
Affiliation(s)
- Gianno Pannafino
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, NY 14853, USA
| | - Jun Jie Chen
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, NY 14853, USA
| | - Viraj Mithani
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, NY 14853, USA
| | - Lisette Payero
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, NY 14853, USA
| | - Michael Gioia
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, NY 14853, USA
| | - J Brooks Crickard
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, NY 14853, USA
| | - Eric Alani
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, NY 14853, USA
| |
Collapse
|
46
|
Biswas L, Tyc KM, Aboelenain M, Sun S, Dundović I, Vukušić K, Liu J, Guo V, Xu M, Scott RT, Tao X, Tolić IM, Xing J, Schindler K. Maternal genetic variants in kinesin motor domains prematurely increase egg aneuploidy. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.07.04.24309950. [PMID: 39006445 PMCID: PMC11245073 DOI: 10.1101/2024.07.04.24309950] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/16/2024]
Abstract
The female reproductive lifespan depends on egg quality, particularly euploidy. Mistakes in meiosis leading to egg aneuploidy are common, but the genetic landscape causing this is not well understood due to limited phenotypic data. We identify genetic determinants of reproductive aging via egg aneuploidy using a biobank of maternal exomes linked with maternal age and embryonic aneuploidy data. We found 404 genes with variants enriched in individuals with high egg aneuploidy rates and implicate kinesin protein family genes in aneuploidy risk. Experimental perturbations showed that motor domain variants in these genes increase aneuploidy in mouse oocytes. A knock-in mouse model validated that a specific variant in kinesin KIF18A accelerates reproductive aging and diminishes fertility. These findings suggest potential non-invasive biomarkers for egg quality, aiding personalized fertility medicine. One sentence summary The study identifies novel genetic determinants of reproductive aging linked to egg aneuploidy by analyzing maternal exomes and demonstrates that variants in kinesin genes, specifically KIF18A , contribute to increased aneuploidy and accelerated reproductive aging, offering potential for personalized fertility medicine.
Collapse
|
47
|
Johnston SE. Understanding the Genetic Basis of Variation in Meiotic Recombination: Past, Present, and Future. Mol Biol Evol 2024; 41:msae112. [PMID: 38959451 PMCID: PMC11221659 DOI: 10.1093/molbev/msae112] [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: 04/17/2024] [Revised: 06/03/2024] [Accepted: 06/05/2024] [Indexed: 07/05/2024] Open
Abstract
Meiotic recombination is a fundamental feature of sexually reproducing species. It is often required for proper chromosome segregation and plays important role in adaptation and the maintenance of genetic diversity. The molecular mechanisms of recombination are remarkably conserved across eukaryotes, yet meiotic genes and proteins show substantial variation in their sequence and function, even between closely related species. Furthermore, the rate and distribution of recombination shows a huge diversity within and between chromosomes, individuals, sexes, populations, and species. This variation has implications for many molecular and evolutionary processes, yet how and why this diversity has evolved is not well understood. A key step in understanding trait evolution is to determine its genetic basis-that is, the number, effect sizes, and distribution of loci underpinning variation. In this perspective, I discuss past and current knowledge on the genetic basis of variation in recombination rate and distribution, explore its evolutionary implications, and present open questions for future research.
Collapse
Affiliation(s)
- Susan E Johnston
- Institute of Ecology and Evolution, School of Biological Sciences, University of Edinburgh, Edinburgh EH9 3FL, UK
| |
Collapse
|
48
|
Zhang J, Tian Y, Xu X, Wang B, Huang Z, Song K, Lou S, Kang J, Zhang N, Li J, Weng J, Liang Y, Ma W. PLD1 promotes spindle assembly and migration through regulating autophagy in mouse oocyte meiosis. Autophagy 2024; 20:1616-1638. [PMID: 38513669 PMCID: PMC11210919 DOI: 10.1080/15548627.2024.2333164] [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] [Revised: 03/09/2024] [Accepted: 03/15/2024] [Indexed: 03/23/2024] Open
Abstract
PLD1 has been implicated in cytoskeletal reorganization and vesicle trafficking in somatic cells; however, its function remains unclear in oocyte meiosis. Herein, we found PLD1 stably expresses in mouse oocytes meiosis, with direct interaction with spindle, RAB11A+ vesicles and macroautophagic/autophagic vacuoles. The genetic or chemical inhibition of PLD1 disturbed MTOC clustering, spindle assembly and its cortical migration, also decreased PtdIns(4,5)P2, phosphorylated CFL1 (p-CFL1 [Ser3]) and ACTR2, and their local distribution on MTOC, spindle and vesicles. Furthermore in PLD1-suppressed oocytes, vesicle size was significantly reduced while F-actin density was dramatically increased in the cytoplasm, the asymmetric distribution of autophagic vacuoles was broken and the whole autophagic process was substantially enhanced, as illustrated with characteristic changes in autophagosomes, autolysosome formation and levels of ATG5, BECN1, LC3-II, SQSTM1 and UB. Exogenous administration of PtdIns(4,5)P2 or overexpression of CFL1 hyperphosphorylation mutant (CFL1S3E) could significantly improve polar MTOC focusing and spindle structure in PLD1-depleted oocytes, whereas overexpression of ACTR2 could rescue not only MTOC clustering, and spindle assembly but also its asymmetric positioning. Interestingly, autophagy activation induced similar defects in spindle structure and positioning; instead, its inhibition alleviated the alterations in PLD1-depleted oocytes, and this was highly attributed to the restored levels of PtdIns(4,5)P2, ACTR2 and p-CFL1 (Ser3). Together, PLD1 promotes spindle assembly and migration in oocyte meiosis, by maintaining rational levels of ACTR2, PtdIns(4,5)P2 and p-CFL1 (Ser3) in a manner of modulating autophagy flux. This study for the first time introduces a unique perspective on autophagic activity and function in oocyte meiotic development.Abbreviations: ACTR2/ARP2: actin related protein 2; ACTR3/ARP3: actin related protein 3; ATG5: autophagy related 5; Baf-A1: bafilomycin A1; BFA: brefeldin A; GAPDH: glyceraldehyde-3-phosphate dehydrogenase; GOLGA2/GM130: golgin A2; GV: germinal vesicle; GVBD: germinal vesicle breakdown; IVM: in vitro maturation; MAP1LC3/LC3: microtubule-associated protein 1 light chain 3; MI: metaphase of meiosis I; MII: metaphase of meiosis II; MO: morpholino; MTOC: microtubule-organizing center; MTOR: mechanistic target of rapamycin kinase; PB1: first polar body; PLA: proximity ligation assay; PLD1: phospholipase D1; PtdIns(4,5)P2/PIP2: phosphatidylinositol 4,5-bisphosphate; RAB11A: RAB11A, member RAS oncogene family; RPS6KB1/S6K1: ribosomal protein S6 kinase B1; SQSTM1/p62: sequestosome 1; TEM: transmission electron microscopy; TUBA/α-tubulin: tubulin alpha; TUBG/γ-tubulin: tubulin gamma; UB: ubiquitin; WASL/N-WASP: WASP like actin nucleation promoting factor.
Collapse
Affiliation(s)
- Jiaqi Zhang
- Department of Histology and Embryology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Ying Tian
- Department of Histology and Embryology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Xiangning Xu
- Department of Histology and Embryology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Bicheng Wang
- Department of Histology and Embryology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Ziqi Huang
- Department of Histology and Embryology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Ke Song
- Department of Human Reproductive Medicine, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing, China
| | - Shuo Lou
- Department of Histology and Embryology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Jingyi Kang
- Department of Histology and Embryology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Ningning Zhang
- Department of Histology and Embryology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Jingyu Li
- Department of Human Reproductive Medicine, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing, China
| | - Jing Weng
- Department of Histology and Embryology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Yuanjing Liang
- Department of Histology and Embryology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Wei Ma
- Department of Histology and Embryology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| |
Collapse
|
49
|
Venu V, Harjunmaa E, Dreau A, Brady S, Absher D, Kingsley DM, Jones FC. Fine-scale contemporary recombination variation and its fitness consequences in adaptively diverging stickleback fish. Nat Ecol Evol 2024; 8:1337-1352. [PMID: 38839849 PMCID: PMC11239493 DOI: 10.1038/s41559-024-02434-4] [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: 08/21/2023] [Accepted: 05/02/2024] [Indexed: 06/07/2024]
Abstract
Despite deep evolutionary conservation, recombination rates vary greatly across the genome and among individuals, sexes and populations. Yet the impact of this variation on adaptively diverging populations is not well understood. Here we characterized fine-scale recombination landscapes in an adaptively divergent pair of marine and freshwater populations of threespine stickleback from River Tyne, Scotland. Through whole-genome sequencing of large nuclear families, we identified the genomic locations of almost 50,000 crossovers and built recombination maps for marine, freshwater and hybrid individuals at a resolution of 3.8 kb. We used these maps to quantify the factors driving variation in recombination rates. We found strong heterochiasmy between sexes but also differences in recombination rates among ecotypes. Hybrids showed evidence of significant recombination suppression in overall map length and in individual loci. Recombination rates were lower not only within individual marine-freshwater-adaptive loci, but also between loci on the same chromosome, suggesting selection on linked gene 'cassettes'. Through temporal sampling along a natural hybrid zone, we found that recombinants showed traits associated with reduced fitness. Our results support predictions that divergence in cis-acting recombination modifiers, whose functions are disrupted in hybrids, may play an important role in maintaining differences among adaptively diverging populations.
Collapse
Affiliation(s)
- Vrinda Venu
- Friedrich Miescher Laboratory of the Max Planck Society, Tübingen, Germany.
- Los Alamos National Laboratory, New Mexico, NM, USA.
| | - Enni Harjunmaa
- Friedrich Miescher Laboratory of the Max Planck Society, Tübingen, Germany
- CeGAT GmbH, Tübingen, Germany
| | - Andreea Dreau
- Friedrich Miescher Laboratory of the Max Planck Society, Tübingen, Germany
- Evotec SE 'Campus Curie', Toulouse, France
| | - Shannon Brady
- Deptartment of Developmental Biology, Stanford University School of Medicine, Stanford, CA, USA
| | - Devin Absher
- HudsonAlpha Institute for Biotechnology, Huntsville, AL, USA
| | - David M Kingsley
- Deptartment of Developmental Biology, Stanford University School of Medicine, Stanford, CA, USA
- Howard Hughes Medical Institute, Chevy Chase, MD, USA
| | - Felicity C Jones
- Friedrich Miescher Laboratory of the Max Planck Society, Tübingen, Germany.
- Groningen Institute for Evolutionary Life Sciences (GELIFES), University of Groningen, Groningen, the Netherlands.
| |
Collapse
|
50
|
Galatidou S, Petelski AA, Pujol A, Lattes K, Latorraca LB, Fair T, Popovic M, Vassena R, Slavov N, Barragán M. Single-cell proteomics reveals decreased abundance of proteostasis and meiosis proteins in advanced maternal age oocytes. Mol Hum Reprod 2024; 30:gaae023. [PMID: 38870523 DOI: 10.1093/molehr/gaae023] [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: 01/16/2024] [Revised: 05/28/2024] [Indexed: 06/15/2024] Open
Abstract
Advanced maternal age is associated with a decline in oocyte quality, which often leads to reproductive failure in humans. However, the mechanisms behind this age-related decline remain unclear. To gain insights into this phenomenon, we applied plexDIA, a multiplexed data-independent acquisition, single-cell mass spectrometry method, to analyze the proteome of oocytes from both young women and women of advanced maternal age. Our findings primarily revealed distinct proteomic profiles between immature fully grown germinal vesicle and mature metaphase II oocytes. Importantly, we further show that a woman's age is associated with changes in her oocyte proteome. Specifically, when compared to oocytes obtained from young women, advanced maternal age oocytes exhibited lower levels of the proteasome and TRiC complex, as well as other key regulators of proteostasis and meiosis. This suggests that aging adversely affects the proteostasis and meiosis networks in human oocytes. The proteins identified in this study hold potential as targets for improving oocyte quality and may guide future studies into the molecular processes underlying oocyte aging.
Collapse
Affiliation(s)
- Styliani Galatidou
- Research and Development, EUGIN Group, Barcelona, Spain
- School of Agriculture and Food Science, University College Dublin, Dublin, Ireland
| | - Aleksandra A Petelski
- Department of Bioengineering, Single Cell Proteomics Center and Barnett Institute, Northeastern University, Boston, MA, USA
| | | | | | - Lais B Latorraca
- School of Agriculture and Food Science, University College Dublin, Dublin, Ireland
| | - Trudee Fair
- School of Agriculture and Food Science, University College Dublin, Dublin, Ireland
| | - Mina Popovic
- Research and Development, EUGIN Group, Barcelona, Spain
| | - Rita Vassena
- Research and Development, EUGIN Group, Barcelona, Spain
| | - Nikolai Slavov
- Department of Bioengineering, Single Cell Proteomics Center and Barnett Institute, Northeastern University, Boston, MA, USA
| | | |
Collapse
|