1
|
Ya A, Deng C, Godek KM. Cell competition eliminates aneuploid human pluripotent stem cells. Stem Cell Reports 2025:102506. [PMID: 40409259 DOI: 10.1016/j.stemcr.2025.102506] [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: 05/23/2024] [Revised: 04/21/2025] [Accepted: 04/23/2025] [Indexed: 05/25/2025] Open
Abstract
Human pluripotent stem cells (hPSCs) maintain diploid populations for generations despite frequent mitotic errors that cause aneuploidy or chromosome imbalances. Consequently, aneuploid hPSC propagation must be prevented to sustain genome stability, but how this is achieved is unknown. Surprisingly, we find that, unlike somatic cells, uniformly aneuploid hPSC populations with heterogeneous abnormal karyotypes proliferate. Instead, in mosaic populations, cell-non-autonomous competition between neighboring diploid and aneuploid hPSCs eliminates less fit aneuploid cells, regardless of specific chromosome imbalances. Aneuploid hPSCs with lower MYC or higher p53 levels relative to diploid neighbors are outcompeted but conversely gain an advantage when MYC and p53 relative abundance switches. Thus, MYC- and p53-driven cell competition preserves hPSC genome integrity despite their low mitotic fidelity and intrinsic capacity to proliferate with an aneuploid genome. These findings have important implications for using hPSCs in regenerative medicine and for how diploid human embryos form during development despite the prevalence of aneuploidy.
Collapse
Affiliation(s)
- Amanda Ya
- Department of Biochemistry and Cell Biology, Geisel School of Medicine at Dartmouth, Hanover, NH, USA; Dartmouth Cancer Center, Geisel School of Medicine at Dartmouth, Lebanon, NH, USA
| | - Chenhui Deng
- Department of Biochemistry and Cell Biology, Geisel School of Medicine at Dartmouth, Hanover, NH, USA; Dartmouth Cancer Center, Geisel School of Medicine at Dartmouth, Lebanon, NH, USA
| | - Kristina M Godek
- Department of Biochemistry and Cell Biology, Geisel School of Medicine at Dartmouth, Hanover, NH, USA; Dartmouth Cancer Center, Geisel School of Medicine at Dartmouth, Lebanon, NH, USA.
| |
Collapse
|
2
|
Cearlock A, Mysliwiec H, Agarsheva M, Krzyspiak J, Ozair MZ, Brivanlou AH, Yang M. Exploring and validating the marmoset as a primate model for chromosomal instability in early development. Mol Hum Reprod 2025; 31:gaaf012. [PMID: 40193493 DOI: 10.1093/molehr/gaaf012] [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: 12/18/2024] [Revised: 03/27/2025] [Indexed: 04/09/2025] Open
Abstract
Aneuploidy in embryos poses a major barrier to successful human reproduction, contributing to nearly 50% of early miscarriages. Despite its high prevalence in human embryos, the molecular mechanisms regulating aneuploid cell fate during development remain poorly understood. This knowledge gap persists due to ethical constraints in human embryo research and the limitations of existing animal models. In this study, we identified the New World primate marmoset (Callithrix jacchus) as a suitable model for investigating aneuploidy. By calling copy number variants from single-cell RNA-sequencing data of marmoset embryonic cells, we identified heterogeneous aneuploidy, indicating chromosomal instability (CIN) in marmoset preimplantation embryos. Furthermore, marmoset aneuploidy displayed lineage-specific behavior during gastruloid differentiation, similar to humans, suggesting a conserved regulatory mechanism in lineage specification. To develop a more pluripotent cell line to study early specification, we established an efficient approach for generating naïve-like marmoset pluripotent stem cells (cjPSCs). These cells resemble preimplantation epiblast-like cells and exhibit inherent CIN. Transcriptome analysis identified potential pathways contributing to aneuploidy during early embryogenesis, including the downregulation of cell cycle checkpoint signaling and the upregulation of autophagy pathways. Additionally, we found no significant effect of spontaneously occurring aneuploidy in cjPSCs on blastoid formation, suggesting that the consequences of aneuploidy become evident only after gastrulation, with preimplantation lineages exhibiting a higher tolerance for genomic instability. Unexpectedly, aneuploidy enhanced cavity formation during blastoid development, suggesting a potential role in facilitating efficient trophectoderm differentiation. Our findings validate the marmoset as a valuable model for studying CIN during early primate development and provide insight into the mechanisms underlying the prevalence of aneuploidy in primates. Naïve-like cjPSCs recapitulate key phenotypic traits of early embryonic cells, providing a robust system for studying post-implantation aneuploid cell fates in vivo and serving as a foundation for future research in this field.
Collapse
Affiliation(s)
- Andrew Cearlock
- Department of Obstetrics & Gynecology, University of Washington, Seattle, WA, USA
- Institute for Stem Cell & Regenerative Medicine, University of Washington, Seattle, WA, USA
| | - Hubert Mysliwiec
- Laboratory of Synthetic Embryology, The Rockefeller University, New York, NY, USA
| | - Margarita Agarsheva
- Laboratory of Synthetic Embryology, The Rockefeller University, New York, NY, USA
| | - Joanna Krzyspiak
- Laboratory of Synthetic Embryology, The Rockefeller University, New York, NY, USA
| | - Mohammad Zeeshan Ozair
- Laboratory of Synthetic Embryology, The Rockefeller University, New York, NY, USA
- Department of Radiation Oncology, Montefiore Einstein Comprehensive Cancer Center, Bronx, New York, NY, USA
| | - Ali H Brivanlou
- Laboratory of Synthetic Embryology, The Rockefeller University, New York, NY, USA
| | - Min Yang
- Department of Obstetrics & Gynecology, University of Washington, Seattle, WA, USA
- Institute for Stem Cell & Regenerative Medicine, University of Washington, Seattle, WA, USA
- Brotman Baty Institute for Precision Medicine, Seattle, WA, USA
- Washington National Primate Center, Seattle, WA, USA
| |
Collapse
|
3
|
De Vos A, De Munck N. Trophectoderm Biopsy: Present State of the Art. Genes (Basel) 2025; 16:134. [PMID: 40004463 PMCID: PMC11854799 DOI: 10.3390/genes16020134] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2024] [Revised: 01/10/2025] [Accepted: 01/20/2025] [Indexed: 02/27/2025] Open
Abstract
Trophectoderm (TE) biopsy is at present the most widely used procedure for preimplantation genetic testing (PGT). At the blastocyst stage, more TE cells (five to seven) can be obtained for genetic analysis. While removing TE cells and not touching the inner cell mass (ICM), the procedure is less invasive. Due to a natural selection happening between day 3 and day 5, 6 or 7 of human embryo development, fewer embryos will have to be biopsied and tested. An additional benefit, especially in view of aneuploidy testing (PGT-A), is the lower level of mosaicism present at the blastocyst stage. The biopsy procedure involves two steps: laser-assisted zona pellucida (ZP) opening and the excision of five to eight TE cells from the blastocyst with or without additional laser energy. Different protocols have emerged over time with variations regarding the technique, the exact moment of ZP opening, and the method of cell removal. The 'pulling' method involves laser excision, whereas the 'flicking' method represents a mechanical approach with or without laser assistance. Embryo developmental speed reaching the full/expanded or hatching/hatched blastocyst stage dictates the timing of the procedure, mostly on day 5 post-insemination, and to a lesser extent on day 6 or even on day 7. The inclusion of lesser quality or delayed blastocysts may impact the quality of the TE sample as well as the clinical outcome. Intracytoplasmic sperm injection (ICSI) is still the preferred method of fertilization for PGT-M (monogenic disorders) and PGT-SR (structural rearrangements). However, conventional in vitro fertilization (IVF) seems feasible for PGT-A (aneuploidy testing). In the absence of a (conclusive) genetic result, the re-biopsy of cryopreserved blastocysts is possible, however, with reduced clinical outcomes. So far, neonatal outcome post-TE biopsy has so far been reassuringly documented.
Collapse
Affiliation(s)
- Anick De Vos
- Brussels IVF, Universitair Ziekenhuis Brussel (UZ Brussel), Laarbeeklaan 101, 1090 Brussels, Belgium;
| | - Neelke De Munck
- Brussels IVF, Universitair Ziekenhuis Brussel (UZ Brussel), Laarbeeklaan 101, 1090 Brussels, Belgium;
- Research Group Genetics, Reproduction and Development (GRAD), Vrije Universiteit Brussel (VUB), Laarbeeklaan 101, 1090 Brussels, Belgium
| |
Collapse
|
4
|
Viville S, Aboulghar M. PGT-A: what's it for, what's wrong? J Assist Reprod Genet 2025; 42:63-69. [PMID: 39847200 PMCID: PMC11806166 DOI: 10.1007/s10815-025-03400-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Accepted: 01/10/2025] [Indexed: 01/24/2025] Open
Abstract
PGT-A, what's it for? Considering the increase in fetal aneuploidies with a woman's age and the high number of miscarriages associated with fetal karyotype anomalies, the concept of selecting IVF embryos based on their karyotype in order to transfer only euploid embryos and eliminate aneuploid ones was proposed. Preimplantation genetic testing for aneuploidy (PGT-A) was then established, nearly 30 years ago, with the expectation that the transfer of euploid embryos would lead to a significant improvement in medically assisted reproduction (MAR) outcomes. PGT-A, what's wrong? Despite the practice and widespread use, PGT-A has not consistently proven its effectiveness. The clinical value of PGT-A remains controversial. The initial studies reported an increase in MAR outcomes. However, these studies used embryo transfer as the reference point. More recent studies, which use intention-to-treat as the reference point, show, at best, slight improvements and, at worst, a reduction in the considered IVF outcomes. In this article, we attempt to answer two key questions: "What is it for?" and "What's wrong with PGT-A?". We also explore some of the ethical issues raised by these conclusions. Ultimately, we suggest that PGT-A should no longer be offered to infertile couples.
Collapse
Affiliation(s)
- Stéphane Viville
- Laboratoire de Diagnostic Génétique, Hôpitaux Universitaires de Strasbourg, UF3472-Génétique de L'infertilité, 67000, Strasbourg, France.
- Institute for Genetics and Molecular and Cellular Biology (IGBMC), University of Strasbourg, CNRS UMR7104, INSERM U1258, Illkirch, France.
| | - Mohamed Aboulghar
- Department of Obstetrics and Gynecology, Cairo University, Cairo, Egypt
- The Egyptian IVF Center, Cairo, Egypt
| |
Collapse
|
5
|
Regin M, Lei Y, Couvreu De Deckersberg E, Janssens C, Huyghebaert A, Guns Y, Verdyck P, Verheyen G, Van de Velde H, Sermon K, Spits C. Complex aneuploidy triggers autophagy and p53-mediated apoptosis and impairs the second lineage segregation in human preimplantation embryos. eLife 2024; 12:RP88916. [PMID: 39652462 PMCID: PMC11627504 DOI: 10.7554/elife.88916] [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] [Indexed: 12/12/2024] Open
Abstract
About 70% of human cleavage stage embryos show chromosomal mosaicism, falling to 20% in blastocysts. Chromosomally mosaic human blastocysts can implant and lead to healthy new-borns with normal karyotypes. Studies in mouse embryos and human gastruloids showed that aneuploid cells are eliminated from the epiblast by p53-mediated apoptosis while being tolerated in the trophectoderm. These observations suggest a selective loss of aneuploid cells from human embryos, but the underlying mechanisms are not yet fully understood. Here, we investigated the cellular consequences of aneuploidy in a total of 125 human blastocysts. RNA-sequencing of trophectoderm cells showed activated p53 pathway and apoptosis proportionate to the level of chromosomal imbalance. Immunostaining corroborated that aneuploidy triggers proteotoxic stress, autophagy, p53-signaling, and apoptosis independent from DNA damage. Total cell numbers were lower in aneuploid embryos, due to a decline both in trophectoderm and in epiblast/primitive endoderm cell numbers. While lower cell numbers in trophectoderm may be attributed to apoptosis, aneuploidy impaired the second lineage segregation, particularly primitive endoderm formation. This might be reinforced by retention of NANOG. Our findings might explain why fully aneuploid embryos fail to further develop and we hypothesize that the same mechanisms lead to the removal of aneuploid cells from mosaic embryos.
Collapse
Affiliation(s)
- Marius Regin
- Brussels Health Campus/Faculty of Medicine and Pharmacy, Research Group Genetics Reproduction and Development, Vrije Universiteit BrusselBrusselsBelgium
| | - Yingnan Lei
- Brussels Health Campus/Faculty of Medicine and Pharmacy, Research Group Genetics Reproduction and Development, Vrije Universiteit BrusselBrusselsBelgium
| | - Edouard Couvreu De Deckersberg
- Brussels Health Campus/Faculty of Medicine and Pharmacy, Research Group Genetics Reproduction and Development, Vrije Universiteit BrusselBrusselsBelgium
| | - Charlotte Janssens
- Brussels Health Campus/Faculty of Medicine and Pharmacy, Research Group Genetics Reproduction and Development, Vrije Universiteit BrusselBrusselsBelgium
| | - Anfien Huyghebaert
- Brussels Health Campus/Faculty of Medicine and Pharmacy, Research Group Genetics Reproduction and Development, Vrije Universiteit BrusselBrusselsBelgium
| | - Yves Guns
- Brussels Health Campus, Brussels IVF, Universitair Ziekenhuis Brussel (UZ Brussel)BrusselsBelgium
| | - Pieter Verdyck
- Brussels Health Campus/Faculty of Medicine and Pharmacy, Research Group Genetics Reproduction and Development, Vrije Universiteit BrusselBrusselsBelgium
- Brussels Health Campus, Medical Genetics, Universitair Ziekenhuis Brussel (UZ Brussel)BrusselsBelgium
| | - Greta Verheyen
- Brussels Health Campus, Brussels IVF, Universitair Ziekenhuis Brussel (UZ Brussel)BrusselsBelgium
| | - Hilde Van de Velde
- Brussels Health Campus/Faculty of Medicine and Pharmacy, Research Group Genetics Reproduction and Development, Vrije Universiteit BrusselBrusselsBelgium
- Brussels Health Campus, Brussels IVF, Universitair Ziekenhuis Brussel (UZ Brussel)BrusselsBelgium
| | - Karen Sermon
- Brussels Health Campus/Faculty of Medicine and Pharmacy, Research Group Genetics Reproduction and Development, Vrije Universiteit BrusselBrusselsBelgium
| | - Claudia Spits
- Brussels Health Campus/Faculty of Medicine and Pharmacy, Research Group Genetics Reproduction and Development, Vrije Universiteit BrusselBrusselsBelgium
| |
Collapse
|
6
|
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
|
7
|
Kósa M, Horváth E, Kalmár T, Maróti Z, Földesi I, Bereczki C. A Patient Diagnosed with Mosaic Trisomy 18 Presenting New Symptoms: Diaphragmatic Relaxation and Cyclic Vomiting Syndrome. Updated Review of Mosaic Trisomy 18 Cases. J Pediatr Genet 2024; 13:320-325. [PMID: 39502855 PMCID: PMC11534437 DOI: 10.1055/s-0042-1757621] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Accepted: 08/31/2022] [Indexed: 12/03/2022]
Abstract
Although data on T18 are widespread, there is a lack of knowledge on mosaic trisomy 18 (mT18). A current review of mT18 symptomatology, long-term follow-up, and potential health risks is lacking for health care professionals. Our paper addresses these, emphasizing the importance of regular tumor screening as a key message for mT18 patient follow-up. We also present the case of a female patient with mT18 who presented with diaphragmatic relaxation and cyclic vomiting syndrome (CVS), which had previously not been reported in this genetic condition. On further investigating the etiology of CVS, we revealed a novel mitochondrial mutation in the MT-ND6 gene in heteroplasmic form. Based on the literature, we hypothesize that the mitochondrial mutation together with mT18 could result in CVS.
Collapse
Affiliation(s)
- Magdolna Kósa
- Department of Pediatrics and Pediatric Health Center, Albert Szent-Györgyi Health Centre, University of Szeged, Szeged, Hungary
| | - Emese Horváth
- Department of Medical Genetics, Albert Szent-Györgyi Health Centre, University of Szeged, Szeged, Hungary
| | - Tibor Kalmár
- Department of Pediatrics and Pediatric Health Center, Albert Szent-Györgyi Health Centre, University of Szeged, Szeged, Hungary
| | - Zoltán Maróti
- Department of Pediatrics and Pediatric Health Center, Albert Szent-Györgyi Health Centre, University of Szeged, Szeged, Hungary
| | - Imre Földesi
- Department of Laboratory Medicine, Albert Szent-Györgyi Health Centre, University of Szeged, Szeged, Hungary
| | - Csaba Bereczki
- Department of Pediatrics and Pediatric Health Center, Albert Szent-Györgyi Health Centre, University of Szeged, Szeged, Hungary
| |
Collapse
|
8
|
Vales JP, Barbaric I. Culture-acquired genetic variation in human pluripotent stem cells: Twenty years on. Bioessays 2024; 46:e2400062. [PMID: 38873900 PMCID: PMC11589660 DOI: 10.1002/bies.202400062] [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: 03/16/2024] [Revised: 06/02/2024] [Accepted: 06/05/2024] [Indexed: 06/15/2024]
Abstract
Genetic changes arising in human pluripotent stem cells (hPSC) upon culture may bestow unwanted or detrimental phenotypes to cells, thus potentially impacting on the applications of hPSCs for clinical use and basic research. In the 20 years since the first report of culture-acquired genetic aberrations in hPSCs, a characteristic spectrum of recurrent aberrations has emerged. The preponderance of such aberrations implies that they provide a selective growth advantage to hPSCs upon expansion. However, understanding the consequences of culture-acquired variants for specific applications in cell therapy or research has been more elusive. The rapid progress of hPSC-based therapies to clinics is galvanizing the field to address this uncertainty and provide definitive ways both for risk assessment of variants and reducing their prevalence in culture. Here, we aim to provide a timely update on almost 20 years of research on this fascinating, but a still unresolved and concerning, phenomenon.
Collapse
Affiliation(s)
- John P. Vales
- Centre for Stem Cell BiologySchool of BiosciencesUniversity of SheffieldSheffieldUK
- Neuroscience InstituteUniversity of SheffieldSheffieldUK
- INSIGNEO InstituteUniversity of SheffieldSheffieldUK
| | - Ivana Barbaric
- Centre for Stem Cell BiologySchool of BiosciencesUniversity of SheffieldSheffieldUK
- Neuroscience InstituteUniversity of SheffieldSheffieldUK
- INSIGNEO InstituteUniversity of SheffieldSheffieldUK
| |
Collapse
|
9
|
Khajehoseini F, Noormohammadi Z, Eftekhari-Yazdi P, Gourabi H, Pazhoomand R, Hosseinishenatal S, Bazrgar M. Evaluation of utilization of amplified blastocoel fluid DNA gel electrophoresis band intensity as an additional minimally invasive approach in embryo selection: A cross-sectional study. Int J Reprod Biomed 2024; 22:907-918. [PMID: 39866585 PMCID: PMC11757668 DOI: 10.18502/ijrm.v22i11.17823] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Revised: 06/05/2024] [Accepted: 10/27/2024] [Indexed: 01/28/2025] Open
Abstract
Background Embryo selection for transfer is critical in assisted reproduction. The presence of DNA in the blastocoel cavity of human blastocysts is assumed to be a consequence of common preimplantation chromosomal abnormalities. Objective This study examined the relationship between the amount of blastocoel fluid (BF) DNA and the band intensity of amplified BF-DNA in gel electrophoresis, considering the influence of ploidy status. Materials and Methods This cross-sectional study categorizes blastocysts into 2 groups based on the array comparative genomic hybridization results by trophectoderm biopsy -the euploid and aneuploid group. After morphological scoring, a biopsy of BF was performed for whole genome amplification, followed by an assessment of band intensity and BF-DNA quantification. The relationship between BF-DNA levels, band intensity, and ploidy status were analyzed. Results The level of BF-DNA was higher in the aneuploid group compared to the euploid group, but the difference was not statistically significant (p = 0.2). We observed that the band intensity was affected by the ploidy status of the embryos, although this difference was not statistically significant (p = 0.059). Notably, electrophoresis band of all embryos with chromosomal loss was strong. No correlation was observed between embryo morphology and chromosomal ploidy (p = 0.8). Conclusion Our findings indicate that BF-DNA band intensity on agarose gel is not currently applicable for preimplantation embryo selection. It seems that embryos with chromosomal loss are more prone to DNA release to BF. The disrelation between embryo morphology and ploidy status represents the necessity of minimally invasive embryo screening methods based on ploidy status.
Collapse
Affiliation(s)
- Fattaneh Khajehoseini
- Department of Biology, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Zahra Noormohammadi
- Department of Biology, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Poopak Eftekhari-Yazdi
- Department of Embryology, Reproductive Biomedicine Research Center, Royan Institute for Reproductive Biomedicine, ACECR, Tehran, Iran
| | - Hamid Gourabi
- Department of Genetics, Reproductive Biomedicine Research Center, Royan Institute for Reproductive Biomedicine, ACECR, Tehran, Iran
| | - Reza Pazhoomand
- Medical Genetics Laboratory, Shiraz Fertility Center, Shiraz, Iran
| | | | - Masood Bazrgar
- Department of Genetics, Reproductive Biomedicine Research Center, Royan Institute for Reproductive Biomedicine, ACECR, Tehran, Iran
| |
Collapse
|
10
|
Alba PG, Inés FB, Julieta Gabriela H, Beatriz GC, Ismael LT, Leopoldo GB, Priscila RI, Pablo BÁ. SMC2 ablation impairs bovine embryo development shortly after blastocyst hatching. Reproduction 2024; 168:e240211. [PMID: 39231091 PMCID: PMC11466199 DOI: 10.1530/rep-24-0211] [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: 06/16/2024] [Accepted: 09/04/2024] [Indexed: 09/06/2024]
Abstract
In brief Bovine embryos lacking SMC2 (a core component of condensins I and II) are unable to survive maternal recognition of pregnancy. SMC2 KO embryos are able to form blastocysts, exhibiting a reduced cell proliferation ability, and arrest their development shortly after hatching. Abstract Condensins are large protein complexes required for chromosome assembly and segregation during mitosis and meiosis. Mouse or bovine embryos lacking SMC2 (a core component of condensins I and II) do not complete development to term, but it is unknown when they arrest their development. Herein, we have assessed the developmental ability of bovine embryos lacking SMC2 due to a naturally occurring mutation termed HH3 (Holstein Haplotype 3) or by CRISPR-mediated gene ablation. To determine if embryos homozygous for the HH3 allele survive to maternal recognition of pregnancy, embryonic day (E)14 embryos were flushed from superovulated carrier cows inseminated with a carrier bull. Mendelian inheritance of the HH3 allele was observed at E14 conceptuses but conceptuses homozygous for HH3 failed to achieve elongation and lacked an embryonic disc. To assess the consequence of the ablation of condensins I and II at earlier developmental stages, SMC2 KO bovine embryos were generated in vitro using CRISPR technology. SMC2 KO embryos were able to form blastocysts but exhibited reduced cell proliferation as evidenced by a significantly lower number of total, trophectoderm (CDX2+), and inner cell mass (SOX2+) cells at Day (D) 8 post-fertilization compared to their WT counterparts and were unable to survive to D12 in vitro. SMC2 ablation did not alter relative telomere length at D8, D12, or E14. In conclusion, condensins I and II are required for blastomere mitosis during early development, and embryos lacking those complexes arrest their development shortly after blastocyst hatching.
Collapse
Affiliation(s)
| | | | - Hamze Julieta Gabriela
- Department of Animal Reproduction, INIA, CSIC, Madrid, Spain
- Department of Cell Biology and Histology, Universidad de Murcia. International Excellence Campus for Higher Education and Research (Campus Mare Nostrum), Murcia, Spain
| | | | | | | | | | | |
Collapse
|
11
|
Shan G, Abdalla K, Liu H, Dai C, Tan J, Law J, Steinberg C, Li A, Kuznyetsova I, Zhang Z, Librach C, Sun Y. Non-invasively predicting euploidy in human blastocysts via quantitative 3D morphology measurement: a retrospective cohort study. Reprod Biol Endocrinol 2024; 22:132. [PMID: 39468586 PMCID: PMC11514912 DOI: 10.1186/s12958-024-01302-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Accepted: 10/17/2024] [Indexed: 10/30/2024] Open
Abstract
BACKGROUND Blastocyst morphology has been demonstrated to be associated with ploidy status. Existing artificial intelligence models use manual grading or 2D images as the input for euploidy prediction, which suffer from subjectivity from observers and information loss due to incomplete features from 2D images. Here we aim to predict euploidy in human blastocysts using quantitative morphological parameters obtained by 3D morphology measurement. METHODS Multi-view images of 226 blastocysts on Day 6 were captured by manually rotating blastocysts during the preparation stage of trophectoderm biopsy. Quantitative morphological parameters were obtained by 3D morphology measurement. Six machine learning models were trained using 3D morphological parameters as the input and PGT-A results as the ground truth outcome. Model performance, including sensitivity, specificity, precision, accuracy and AUC, was evaluated on an additional test dataset. Model interpretation was conducted on the best-performing model. RESULTS All the 3D morphological parameters were significantly different between euploid and non-euploid blastocysts. Multivariate analysis revealed that three of the five parameters including trophectoderm cell number, trophectoderm cell size variance and inner cell mass area maintained statistical significance (P < 0.001, aOR = 1.054, 95% CI 1.034-1.073; P = 0.003, aOR = 0.994, 95% CI 0.991-0.998; P = 0.010, aOR = 1.003, 95% CI 1.001-1.006). The accuracy of euploidy prediction by the six machine learning models ranged from 80 to 95.6%, and the AUCs ranged from 0.881 to 0.984. Particularly, the decision tree model achieved the highest accuracy of 95.6% (95% CI 84.9-99.5%) with the AUC of 0.978 (95% CI 0.882-0.999), and the extreme gradient boosting model achieved the highest AUC of 0.984 (95% CI 0.892-1.000) with the accuracy of 93.3% (95% CI 81.7-98.6%). No significant difference was found between different age groups using either decision tree or extreme gradient boosting to predict euploid blastocysts. The quantitative criteria extracted from the decision tree imply that euploid blastocysts have a higher number of trophectoderm cells, larger inner cell mass area, and smaller trophectoderm cell size variance compared to non-euploid blastocysts. CONCLUSIONS Using quantitative morphological parameters obtained by 3D morphology measurement, the decision tree-based machine learning model achieved an accuracy of 95.6% and AUC of 0.978 for predicting euploidy in Day 6 human blastocysts. TRIAL REGISTRATION N/A.
Collapse
Affiliation(s)
- Guanqiao Shan
- Department of Mechanical and Industrial Engineering, University of Toronto, Toronto, ON, M5S 3G8, Canada
| | - Khaled Abdalla
- Department of Mechanical and Industrial Engineering, University of Toronto, Toronto, ON, M5S 3G8, Canada
| | - Hang Liu
- Department of Mechanical and Industrial Engineering, University of Toronto, Toronto, ON, M5S 3G8, Canada
| | - Changsheng Dai
- School of Mechanical Engineering, Dalian University of Technology, Dalian, 116024, China
| | - Justin Tan
- CReATe Fertility Centre, Toronto, ON, M5G 1N8, Canada
| | - Junhui Law
- Department of Mechanical and Industrial Engineering, University of Toronto, Toronto, ON, M5S 3G8, Canada
| | | | - Ang Li
- Department of Computer Science, University of Toronto, Toronto, ON, M5S 2E4, Canada
| | | | - Zhuoran Zhang
- School of Science and Engineering, The Chinese University of Hong Kong Shenzhen, Shenzhen, 518172, China.
| | - Clifford Librach
- CReATe Fertility Centre, Toronto, ON, M5G 1N8, Canada.
- Department of Obstetrics and Gynecology, University of Toronto, Toronto, ON, M5G 1E2, Canada.
- Sunnybrook Research Institute, Toronto, ON, M4N 3M5, Canada.
| | - Yu Sun
- Department of Mechanical and Industrial Engineering, University of Toronto, Toronto, ON, M5S 3G8, Canada.
- Department of Computer Science, University of Toronto, Toronto, ON, M5S 2E4, Canada.
| |
Collapse
|
12
|
Shao DD, Zhao Y, Ghosh U, Brew J, Zhao S, Qian X, Tran J, Taketomi T, Tsuruta F, Park PJ, Walsh CA. Perinatal Reduction of Genetically Aberrant Neurons from Human Cerebral Cortex. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.08.617159. [PMID: 39416114 PMCID: PMC11482944 DOI: 10.1101/2024.10.08.617159] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/19/2024]
Abstract
Since human neurons are postmitotic and long-lived, the regulation of their genomic content is crucial. Normal neuronal function is uniquely dependent on gene dosage, with diverse genome copy number alterations (CNA) associated with neurodevelopmental and neuropsychiatric conditions 1-3 . In this study, we evaluated the landscape of CNA arising in normal human brains, focusing on prenatal and perinatal ages. We surveyed ∼5,897 CNA in >1,200 single neurons from human postmortem brain of individuals without a neurological diagnosis, ranging in age from gestational week (GW) 14 to 90 years old. Using Tn5-based single-cell whole-genome amplification (scWGA) and informatic advances to validate CNAs in single neurons, we determined that a striking proportion of neurons (up to 45%) in human prenatal cortex showed aberrant genomes, characterized by large-scale CNAs in multiple chromosomes, which reduces significantly during the perinatal period (p<0.1). Furthermore, we identified micronuclei in the developing cortex, reflecting genetic instability reminiscent of that described in early embryonic development 4-6 . The scale of CNA appeared to alter the trajectory of neuronal elimination, as subchromosomal CNAs were more slowly eliminated, over the course of a lifetime. CNAs were depleted for dosage-sensitive genes and genes involved in neurodevelopmental disorders (p<.05), and thus represent genomic quality control mechanisms that eliminate selectively those neurons with CNA involving critical genes. Perinatal elimination of defective neuronal genomes may in turn reflect a developmental landmark essential for normal cognitive function.
Collapse
|
13
|
Shahbazi MN, Pasque V. Early human development and stem cell-based human embryo models. Cell Stem Cell 2024; 31:1398-1418. [PMID: 39366361 PMCID: PMC7617107 DOI: 10.1016/j.stem.2024.09.002] [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: 03/15/2024] [Revised: 06/18/2024] [Accepted: 09/02/2024] [Indexed: 10/06/2024]
Abstract
The use of stem cells to model the early human embryo promises to transform our understanding of developmental biology and human reproduction. In this review, we present our current knowledge of the first 2 weeks of human embryo development. We first focus on the distinct cell lineages of the embryo and the derivation of stem cell lines. We then discuss the intercellular crosstalk that guides early embryo development and how this crosstalk is recapitulated in vitro to generate stem cell-based embryo models. We highlight advances in this fast-developing field, discuss current limitations, and provide a vision for the future.
Collapse
Affiliation(s)
| | - Vincent Pasque
- Department of Development and Regeneration, KU Leuven, 3000 Leuven, Belgium; Leuven Stem Cell Institute & Leuven Institute for Single-Cell Omics (LISCO), Leuven, Belgium.
| |
Collapse
|
14
|
Zhai F, Kong S, Song S, Guo Q, Ding L, Zhang J, Wang N, Kuo Y, Guan S, Yuan P, Yan L, Yan Z, Qiao J. Human embryos harbor complex mosaicism with broad presence of aneuploid cells during early development. Cell Discov 2024; 10:98. [PMID: 39313513 PMCID: PMC11420220 DOI: 10.1038/s41421-024-00719-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Accepted: 07/23/2024] [Indexed: 09/25/2024] Open
Abstract
Pre-implantation genetic testing for aneuploidy (PGT-A) is used in approximately half of in vitro fertilization cycles. Given the limited understanding of the genetics of human embryos, the current use of PGT-A is based on biologically uncertain assumptions and unvalidated guidelines, leading to the possibility of disposing of embryos with pregnancy potential. We isolated and sequenced all single cells (1133) from in vitro cultured 20 human blastocysts. We found that all blastocysts exhibited mosaicism with mitotic-induced aneuploid cells and showed an ~25% aneuploidy rate per embryo. Moreover, 70% (14/20) of blastocysts contained 'chromosome-complementary' cells, suggesting genetic mosaicism is underestimated in routine PGT-A. Additionally, the analysis of 20,945 single cells from day 8-14 embryos (in vitro cultured) and embryonic/fetal organs showed that 97% of the analyzed embryos/organs were mosaic. Over 96% of their aneuploid cells harbored ≤ 2 chromosome errors. Our findings have revealed a high prevalence of mosaicism in human embryos.
Collapse
Affiliation(s)
- Fan Zhai
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China
- National Clinical Research Center for Obstetrics and Gynecology, Beijing, China
- Key Laboratory of Assisted Reproduction (Peking University), Ministry of Education, Beijing, China
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing, China
- Research Units of Comprehensive Diagnosis and Treatment of Oocyte Maturation Arrest, Beijing, China
| | - Siming Kong
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China
- National Clinical Research Center for Obstetrics and Gynecology, Beijing, China
- Key Laboratory of Assisted Reproduction (Peking University), Ministry of Education, Beijing, China
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing, China
- Research Units of Comprehensive Diagnosis and Treatment of Oocyte Maturation Arrest, Beijing, China
- Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, China
| | - Shi Song
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China
- National Clinical Research Center for Obstetrics and Gynecology, Beijing, China
- Key Laboratory of Assisted Reproduction (Peking University), Ministry of Education, Beijing, China
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing, China
- Research Units of Comprehensive Diagnosis and Treatment of Oocyte Maturation Arrest, Beijing, China
| | - Qianying Guo
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China
- National Clinical Research Center for Obstetrics and Gynecology, Beijing, China
- Key Laboratory of Assisted Reproduction (Peking University), Ministry of Education, Beijing, China
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing, China
- Research Units of Comprehensive Diagnosis and Treatment of Oocyte Maturation Arrest, Beijing, China
| | - Ling Ding
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China
- National Clinical Research Center for Obstetrics and Gynecology, Beijing, China
- Key Laboratory of Assisted Reproduction (Peking University), Ministry of Education, Beijing, China
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing, China
- Research Units of Comprehensive Diagnosis and Treatment of Oocyte Maturation Arrest, Beijing, China
| | - Jiaqi Zhang
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China
- National Clinical Research Center for Obstetrics and Gynecology, Beijing, China
- Key Laboratory of Assisted Reproduction (Peking University), Ministry of Education, Beijing, China
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing, China
- Research Units of Comprehensive Diagnosis and Treatment of Oocyte Maturation Arrest, Beijing, China
| | - Nan Wang
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China
- National Clinical Research Center for Obstetrics and Gynecology, Beijing, China
- Key Laboratory of Assisted Reproduction (Peking University), Ministry of Education, Beijing, China
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing, China
- Research Units of Comprehensive Diagnosis and Treatment of Oocyte Maturation Arrest, Beijing, China
| | - Ying Kuo
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China
- National Clinical Research Center for Obstetrics and Gynecology, Beijing, China
- Key Laboratory of Assisted Reproduction (Peking University), Ministry of Education, Beijing, China
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing, China
- Research Units of Comprehensive Diagnosis and Treatment of Oocyte Maturation Arrest, Beijing, China
| | - Shuo Guan
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China
- National Clinical Research Center for Obstetrics and Gynecology, Beijing, China
- Key Laboratory of Assisted Reproduction (Peking University), Ministry of Education, Beijing, China
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing, China
- Research Units of Comprehensive Diagnosis and Treatment of Oocyte Maturation Arrest, Beijing, China
| | - Peng Yuan
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China.
- National Clinical Research Center for Obstetrics and Gynecology, Beijing, China.
- Key Laboratory of Assisted Reproduction (Peking University), Ministry of Education, Beijing, China.
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing, China.
- Research Units of Comprehensive Diagnosis and Treatment of Oocyte Maturation Arrest, Beijing, China.
| | - Liying Yan
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China.
- National Clinical Research Center for Obstetrics and Gynecology, Beijing, China.
- Key Laboratory of Assisted Reproduction (Peking University), Ministry of Education, Beijing, China.
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing, China.
- Research Units of Comprehensive Diagnosis and Treatment of Oocyte Maturation Arrest, Beijing, China.
| | - Zhiqiang Yan
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China.
- National Clinical Research Center for Obstetrics and Gynecology, Beijing, China.
- Key Laboratory of Assisted Reproduction (Peking University), Ministry of Education, Beijing, China.
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing, China.
- Research Units of Comprehensive Diagnosis and Treatment of Oocyte Maturation Arrest, Beijing, China.
| | - Jie Qiao
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China.
- National Clinical Research Center for Obstetrics and Gynecology, Beijing, China.
- Key Laboratory of Assisted Reproduction (Peking University), Ministry of Education, Beijing, China.
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing, China.
- Research Units of Comprehensive Diagnosis and Treatment of Oocyte Maturation Arrest, Beijing, China.
- Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, China.
- Beijing Advanced Innovation Center for Genomics, Beijing, China.
- Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, China.
| |
Collapse
|
15
|
Sran S, Ringland A, Bedrosian TA. Building the brain mosaic: an expanded view. Trends Genet 2024; 40:747-756. [PMID: 38853120 PMCID: PMC11387136 DOI: 10.1016/j.tig.2024.05.008] [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: 03/31/2024] [Revised: 05/20/2024] [Accepted: 05/21/2024] [Indexed: 06/11/2024]
Abstract
The complexity of the brain is closely tied to its nature as a genetic mosaic, wherein each cell is distinguished by a unique constellation of somatic variants that contribute to functional and phenotypic diversity. Postzygotic variation arising during neurogenesis is recognized as a key contributor to brain mosaicism; however, recent advances have broadened our understanding to include sources of neural genomic diversity that develop throughout the entire lifespan, from embryogenesis through aging. Moving beyond the traditional confines of neurodevelopment, in this review, we delve into the complex mechanisms that enable various origins of brain mosaicism.
Collapse
Affiliation(s)
- Sahibjot Sran
- Institute for Genomic Medicine, Nationwide Children's Hospital, Columbus, OH, USA
| | - Amanda Ringland
- Institute for Genomic Medicine, Nationwide Children's Hospital, Columbus, OH, USA
| | - Tracy A Bedrosian
- Institute for Genomic Medicine, Nationwide Children's Hospital, Columbus, OH, USA; Department of Pediatrics, The Ohio State University College of Medicine, Columbus, OH, USA.
| |
Collapse
|
16
|
de Jaime-Soguero A, Hattemer J, Bufe A, Haas A, van den Berg J, van Batenburg V, Das B, di Marco B, Androulaki S, Böhly N, Landry JJM, Schoell B, Rosa VS, Villacorta L, Baskan Y, Trapp M, Benes V, Chabes A, Shahbazi M, Jauch A, Engel U, Patrizi A, Sotillo R, van Oudenaarden A, Bageritz J, Alfonso J, Bastians H, Acebrón SP. Developmental signals control chromosome segregation fidelity during pluripotency and neurogenesis by modulating replicative stress. Nat Commun 2024; 15:7404. [PMID: 39191776 DOI: 10.1038/s41467-024-51821-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Accepted: 08/09/2024] [Indexed: 08/29/2024] Open
Abstract
Human development relies on the correct replication, maintenance and segregation of our genetic blueprints. How these processes are monitored across embryonic lineages, and why genomic mosaicism varies during development remain unknown. Using pluripotent stem cells, we identify that several patterning signals-including WNT, BMP, and FGF-converge into the modulation of DNA replication stress and damage during S-phase, which in turn controls chromosome segregation fidelity in mitosis. We show that the WNT and BMP signals protect from excessive origin firing, DNA damage and chromosome missegregation derived from stalled forks in pluripotency. Cell signalling control of chromosome segregation declines during lineage specification into the three germ layers, but re-emerges in neural progenitors. In particular, we find that the neurogenic factor FGF2 induces DNA replication stress-mediated chromosome missegregation during the onset of neurogenesis, which could provide a rationale for the elevated chromosomal mosaicism of the developing brain. Our results highlight roles for morphogens and cellular identity in genome maintenance that contribute to somatic mosaicism during mammalian development.
Collapse
Affiliation(s)
| | - Janina Hattemer
- Centre for Organismal Studies (COS), Heidelberg University, Heidelberg, Germany
| | - Anja Bufe
- Centre for Organismal Studies (COS), Heidelberg University, Heidelberg, Germany
| | - Alexander Haas
- Department of Molecular Oncology, Section for Cellular Oncology, University Medical Center Göttingen (UMG), Göttingen, Germany
| | - Jeroen van den Berg
- Oncode Institute, Utrecht, The Netherlands
- Hubrecht Institute, Utrecht, The Netherlands
- KNAW (Royal Netherlands Academy of Arts and Sciences), Utrecht, The Netherlands
- University Medical Center Utrecht, Utrecht, The Netherlands
| | - Vincent van Batenburg
- Oncode Institute, Utrecht, The Netherlands
- Hubrecht Institute, Utrecht, The Netherlands
- KNAW (Royal Netherlands Academy of Arts and Sciences), Utrecht, The Netherlands
- University Medical Center Utrecht, Utrecht, The Netherlands
| | - Biswajit Das
- Department of Medical Biochemistry and Biophysics, Umeå University, Umeå, Sweden
| | - Barbara di Marco
- Department of Clinical Neurobiology, University Hospital Heidelberg and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Stefania Androulaki
- Centre for Organismal Studies (COS), Heidelberg University, Heidelberg, Germany
| | - Nicolas Böhly
- Department of Molecular Oncology, Section for Cellular Oncology, University Medical Center Göttingen (UMG), Göttingen, Germany
| | - Jonathan J M Landry
- Genomics Core Facility, European Molecular Biology Laboratory (EMBL), Heidelberg, Germany
| | - Brigitte Schoell
- Institute of Human Genetics, Heidelberg University, Heidelberg, Germany
| | | | - Laura Villacorta
- Genomics Core Facility, European Molecular Biology Laboratory (EMBL), Heidelberg, Germany
| | - Yagmur Baskan
- Centre for Organismal Studies (COS), Heidelberg University, Heidelberg, Germany
| | - Marleen Trapp
- Schaller Research Group, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Vladimir Benes
- Genomics Core Facility, European Molecular Biology Laboratory (EMBL), Heidelberg, Germany
| | - Andrei Chabes
- Department of Medical Biochemistry and Biophysics, Umeå University, Umeå, Sweden
| | | | - Anna Jauch
- Institute of Human Genetics, Heidelberg University, Heidelberg, Germany
| | - Ulrike Engel
- Nikon Imaging Center at the University of Heidelberg, Bioquant, Heidelberg, Germany
| | - Annarita Patrizi
- Schaller Research Group, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Rocio Sotillo
- Division of Molecular Thoracic Oncology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Alexander van Oudenaarden
- Oncode Institute, Utrecht, The Netherlands
- Hubrecht Institute, Utrecht, The Netherlands
- KNAW (Royal Netherlands Academy of Arts and Sciences), Utrecht, The Netherlands
- University Medical Center Utrecht, Utrecht, The Netherlands
| | - Josephine Bageritz
- Centre for Organismal Studies (COS), Heidelberg University, Heidelberg, Germany
| | - Julieta Alfonso
- Department of Clinical Neurobiology, University Hospital Heidelberg and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Holger Bastians
- Department of Molecular Oncology, Section for Cellular Oncology, University Medical Center Göttingen (UMG), Göttingen, Germany
| | - Sergio P Acebrón
- Centre for Organismal Studies (COS), Heidelberg University, Heidelberg, Germany.
| |
Collapse
|
17
|
Sanchez-Vasquez E, Bronner ME, Zernicka-Goetz M. HIF1A contributes to the survival of aneuploid and mosaic pre-implantation embryos. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.09.04.556218. [PMID: 39071426 PMCID: PMC11275769 DOI: 10.1101/2023.09.04.556218] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/30/2024]
Abstract
Human fertility is suboptimal, partly due to error-prone divisions in early cleavage-stages that result in aneuploidy. Most human pre-implantation are mosaics of euploid and aneuploid cells, however, mosaic embryos with a low proportion of aneuploid cells have a similar likelihood of developing to term as fully euploid embryos. How embryos manage aneuploidy during development is poorly understood. This knowledge is crucial for improving fertility treatments and reducing developmental defects. To explore these mechanisms, we established a new mouse model of chromosome mosaicism to study the fate of aneuploid cells during pre-implantation development. We previously used the Mps1 inhibitor reversine to generate aneuploidy in embryos. Here, we found that treatment with the more specific Mps1 inhibitor AZ3146 induced chromosome segregation defects in pre-implantation embryos, similar to reversine. However, AZ3146-treated embryos showed a higher developmental potential than reversine-treated embryos. Unlike reversine-treated embryos, AZ3146-treated embryos exhibited transient upregulation of Hypoxia Inducible-Factor-1A (HIF1A) and lacked p53 upregulation. Pre-implantation embryos develop in a hypoxic environment in vivo, and hypoxia exposure in vitro reduced DNA damage in response to Mps1 inhibition and increased the proportion of euploid cells in the mosaic epiblast. Inhibiting HIF1A in mosaic embryos also decreased the proportion of aneuploid cells in mosaic embryos. Our work illuminates potential strategies to improve the developmental potential of mosaic embryos.
Collapse
Affiliation(s)
| | - Marianne E. Bronner
- Division of Biology 139-74, California Institute of Technology, Pasadena, CA 91125, USA
| | - Magdalena Zernicka-Goetz
- Division of Biology 139-74, California Institute of Technology, Pasadena, CA 91125, USA
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
| |
Collapse
|
18
|
Wang S, Leng L, Wang Q, Gu Y, Li J, An Y, Deng Q, Xie P, Cheng C, Chen X, Zhou Q, Lu J, Chen F, Liu L, Yang H, Wang J, Xu X, Hou Y, Gong F, Hu L, Lu G, Shang Z, Lin G. A single-cell transcriptome atlas of human euploid and aneuploid blastocysts. Nat Genet 2024; 56:1468-1481. [PMID: 38839885 DOI: 10.1038/s41588-024-01788-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Accepted: 05/06/2024] [Indexed: 06/07/2024]
Abstract
Aneuploidy is frequently detected in early human embryos as a major cause of early pregnancy failure. However, how aneuploidy affects cellular function remains elusive. Here, we profiled the transcriptomes of 14,908 single cells from 203 human euploid and aneuploid blastocysts involving autosomal and sex chromosomes. Nearly all of the blastocysts contained four lineages. In aneuploid chromosomes, 19.5% ± 1.2% of the expressed genes showed a dosage effect, and 90 dosage-sensitive domains were identified. Aneuploidy leads to prevalent genome-wide transcriptome alterations. Common effects, including apoptosis, were identified, especially in monosomies, partially explaining the lower cell numbers in autosomal monosomies. We further identified lineage-specific effects causing unstable epiblast development in aneuploidies, which was accompanied by the downregulation of TGF-β and FGF signaling, which resulted in insufficient trophectoderm maturation. Our work provides crucial insights into the molecular basis of human aneuploid blastocysts and may shed light on the cellular interaction during blastocyst development.
Collapse
Affiliation(s)
- Shengpeng Wang
- BGI Research, Hangzhou, China
- BGI Research, Shenzhen, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Lizhi Leng
- Institute of Reproductive and Stem Cell Engineering, School of Basic Medical Science, Central South University, Changsha, China
- NHC Key Laboratory of Human Stem Cell and Reproductive Engineering, Changsha, China
- Reproductive & Genetic Hospital of CITIC-Xiangya, Changsha, China
- Clinical Research Center for Reproduction and Genetics in Hunan Province, Changsha, China
| | | | - Yifan Gu
- Institute of Reproductive and Stem Cell Engineering, School of Basic Medical Science, Central South University, Changsha, China
- NHC Key Laboratory of Human Stem Cell and Reproductive Engineering, Changsha, China
- Reproductive & Genetic Hospital of CITIC-Xiangya, Changsha, China
- Clinical Research Center for Reproduction and Genetics in Hunan Province, Changsha, China
| | | | | | - Qiuting Deng
- BGI Research, Hangzhou, China
- BGI Research, Shenzhen, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Pingyuan Xie
- Hunan Normal University School of Medicine, Changsha, China
- National Engineering and Research Center of Human Stem Cell, Changsha, China
| | - Can Cheng
- Institute of Reproductive and Stem Cell Engineering, School of Basic Medical Science, Central South University, Changsha, China
- NHC Key Laboratory of Human Stem Cell and Reproductive Engineering, Changsha, China
| | - Xueqin Chen
- Institute of Reproductive and Stem Cell Engineering, School of Basic Medical Science, Central South University, Changsha, China
- NHC Key Laboratory of Human Stem Cell and Reproductive Engineering, Changsha, China
| | - Qinwei Zhou
- Reproductive & Genetic Hospital of CITIC-Xiangya, Changsha, China
- Clinical Research Center for Reproduction and Genetics in Hunan Province, Changsha, China
| | - Jia Lu
- Institute of Reproductive and Stem Cell Engineering, School of Basic Medical Science, Central South University, Changsha, China
- NHC Key Laboratory of Human Stem Cell and Reproductive Engineering, Changsha, China
| | - Fang Chen
- BGI Research, Shenzhen, China
- Shenzhen Engineering Laboratory for Birth Defects Screening, BGI Research, Shenzhen, China
| | - Longqi Liu
- BGI Research, Hangzhou, China
- BGI Research, Shenzhen, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Huanming Yang
- BGI Research, Shenzhen, China
- James D. Watson Institute of Genome Science, Hangzhou, China
| | - Jian Wang
- BGI Research, Shenzhen, China
- James D. Watson Institute of Genome Science, Hangzhou, China
| | - Xun Xu
- BGI Research, Hangzhou, China
- BGI Research, Shenzhen, China
- Guangdong Provincial Key Laboratory of Genome Read and Write, Shenzhen, China
| | - Yong Hou
- BGI Research, Shenzhen, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Fei Gong
- Institute of Reproductive and Stem Cell Engineering, School of Basic Medical Science, Central South University, Changsha, China
- NHC Key Laboratory of Human Stem Cell and Reproductive Engineering, Changsha, China
- Reproductive & Genetic Hospital of CITIC-Xiangya, Changsha, China
- Clinical Research Center for Reproduction and Genetics in Hunan Province, Changsha, China
| | - Liang Hu
- Institute of Reproductive and Stem Cell Engineering, School of Basic Medical Science, Central South University, Changsha, China
- NHC Key Laboratory of Human Stem Cell and Reproductive Engineering, Changsha, China
- Reproductive & Genetic Hospital of CITIC-Xiangya, Changsha, China
- Clinical Research Center for Reproduction and Genetics in Hunan Province, Changsha, China
| | - Guangxiu Lu
- Institute of Reproductive and Stem Cell Engineering, School of Basic Medical Science, Central South University, Changsha, China
- NHC Key Laboratory of Human Stem Cell and Reproductive Engineering, Changsha, China
- Reproductive & Genetic Hospital of CITIC-Xiangya, Changsha, China
- Clinical Research Center for Reproduction and Genetics in Hunan Province, Changsha, China
- National Engineering and Research Center of Human Stem Cell, Changsha, China
| | - Zhouchun Shang
- BGI Research, Shenzhen, China.
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China.
| | - Ge Lin
- Institute of Reproductive and Stem Cell Engineering, School of Basic Medical Science, Central South University, Changsha, China.
- NHC Key Laboratory of Human Stem Cell and Reproductive Engineering, Changsha, China.
- Reproductive & Genetic Hospital of CITIC-Xiangya, Changsha, China.
- Clinical Research Center for Reproduction and Genetics in Hunan Province, Changsha, China.
- National Engineering and Research Center of Human Stem Cell, Changsha, China.
| |
Collapse
|
19
|
Gualtieri R, De Gregorio V, Candela A, Travaglione A, Genovese V, Barbato V, Talevi R. In Vitro Culture of Mammalian Embryos: Is There Room for Improvement? Cells 2024; 13:996. [PMID: 38920627 PMCID: PMC11202082 DOI: 10.3390/cells13120996] [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: 04/30/2024] [Revised: 06/03/2024] [Accepted: 06/05/2024] [Indexed: 06/27/2024] Open
Abstract
Preimplantation embryo culture, pivotal in assisted reproductive technology (ART), has lagged in innovation compared to embryo selection advancements. This review examines the persisting gap between in vivo and in vitro embryo development, emphasizing the need for improved culture conditions. While in humans this gap is hardly estimated, animal models, particularly bovines, reveal clear disparities in developmental competence, cryotolerance, pregnancy and live birth rates between in vitro-produced (IVP) and in vivo-derived (IVD) embryos. Molecular analyses unveil distinct differences in morphology, metabolism, and genomic stability, underscoring the need for refining culture conditions for better ART outcomes. To this end, a deeper comprehension of oviduct physiology and embryo transport is crucial for grasping embryo-maternal interactions' mechanisms. Research on autocrine and paracrine factors, and extracellular vesicles in embryo-maternal tract interactions, elucidates vital communication networks for successful implantation and pregnancy. In vitro, confinement, and embryo density are key factors to boost embryo development. Advanced dynamic culture systems mimicking fluid mechanical stimulation in the oviduct, through vibration, tilting, and microfluidic methods, and the use of innovative softer substrates, hold promise for optimizing in vitro embryo development.
Collapse
Affiliation(s)
- Roberto Gualtieri
- Department of Biology, University of Naples ‘’Federico II’’, Complesso Universitario Di Monte S. Angelo, Via Cinthia, 80126 Naples, Italy; (V.D.G.); (A.C.); (A.T.); (V.G.); (V.B.); (R.T.)
| | | | | | | | | | | | | |
Collapse
|
20
|
Ya A, Deng C, Godek KM. Cell Competition Eliminates Aneuploid Human Pluripotent Stem Cells. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.08.593217. [PMID: 38766106 PMCID: PMC11100710 DOI: 10.1101/2024.05.08.593217] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 05/22/2024]
Abstract
Human pluripotent stem cells (hPSCs) maintain diploid populations for generations despite a persistently high rate of mitotic errors that cause aneuploidy, or chromosome imbalances. Consequently, to maintain genome stability, aneuploidy must inhibit hPSC proliferation, but the mechanisms are unknown. Here, we surprisingly find that homogeneous aneuploid populations of hPSCs proliferate unlike aneuploid non-transformed somatic cells. Instead, in mosaic populations, cell non-autonomous competition between neighboring diploid and aneuploid hPSCs eliminates less fit aneuploid cells. Aneuploid hPSCs with lower Myc or higher p53 levels relative to diploid neighbors are outcompeted but conversely gain a selective advantage when Myc and p53 relative abundance switches. Thus, although hPSCs frequently missegregate chromosomes and inherently tolerate aneuploidy, Myc- and p53-driven cell competition preserves their genome integrity. These findings have important implications for the use of hPSCs in regenerative medicine and for how diploid human embryos are established despite the prevalence of aneuploidy during early development.
Collapse
Affiliation(s)
- Amanda Ya
- Department of Biochemistry and Cell Biology, Geisel School of Medicine at Dartmouth, Hanover, NH, USA
- Dartmouth Cancer Center, Geisel School of Medicine at Dartmouth, Lebanon, NH, USA
| | - Chenhui Deng
- Department of Biochemistry and Cell Biology, Geisel School of Medicine at Dartmouth, Hanover, NH, USA
- Dartmouth Cancer Center, Geisel School of Medicine at Dartmouth, Lebanon, NH, USA
| | - Kristina M. Godek
- Department of Biochemistry and Cell Biology, Geisel School of Medicine at Dartmouth, Hanover, NH, USA
- Dartmouth Cancer Center, Geisel School of Medicine at Dartmouth, Lebanon, NH, USA
- Lead contact
| |
Collapse
|
21
|
Zhang X, Zheng PS. Mechanism of chromosomal mosaicism in preimplantation embryos and its effect on embryo development. J Assist Reprod Genet 2024; 41:1127-1141. [PMID: 38386118 PMCID: PMC11143108 DOI: 10.1007/s10815-024-03048-2] [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: 01/30/2024] [Indexed: 02/23/2024] Open
Abstract
Aneuploidy is one of the main causes of miscarriage and in vitro fertilization failure. Mitotic abnormalities in preimplantation embryos are the main cause of mosaicism, which may be influenced by several endogenous factors such as relaxation of cell cycle control mechanisms, defects in chromosome cohesion, centrosome aberrations and abnormal spindle assembly, and DNA replication stress. In addition, incomplete trisomy rescue is a rare cause of mosaicism. However, there may be a self-correcting mechanism in mosaic embryos, which allows some mosaicisms to potentially develop into normal embryos. At present, it is difficult to accurately diagnose mosaicism using preimplantation genetic testing for aneuploidy. Therefore, in clinical practice, embryos diagnosed as mosaic should be considered comprehensively based on the specific situation of the patient.
Collapse
Affiliation(s)
- Xue Zhang
- Department of Reproductive Medicine, The First Affiliated Hospital, Xi'an Jiaotong University of Medical School, Xi'an, 710061, Shanxi, P.R. China
| | - Peng-Sheng Zheng
- Department of Reproductive Medicine, The First Affiliated Hospital, Xi'an Jiaotong University of Medical School, Xi'an, 710061, Shanxi, P.R. China.
- Section of Cancer Stem Cell Research, Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education of People's Republic of China, Xi'an, 710061, Shanxi, P.R. China.
| |
Collapse
|
22
|
Martín Á, Mercader A, Beltrán D, Mifsud A, Nohales M, Pardiñas ML, Ortega-Jaén D, de Los Santos MJ. Trophectoderm cells of human mosaic embryos display increased apoptotic levels and impaired differentiation capacity: a molecular clue regarding their reproductive fate? Hum Reprod 2024; 39:709-723. [PMID: 38308811 DOI: 10.1093/humrep/deae009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Revised: 11/29/2023] [Indexed: 02/05/2024] Open
Abstract
STUDY QUESTION Are there cell lineage-related differences in the apoptotic rates and differentiation capacity of human blastocysts diagnosed as euploid, mosaic, and aneuploid after preimplantation genetic testing for aneuploidy (PGT-A) based on concurrent copy number and genotyping analysis? SUMMARY ANSWER Trophectoderm (TE) cells of mosaic and aneuploid blastocysts exhibit significantly higher levels of apoptosis and significantly reduced differentiation capacity compared to those of euploid blastocysts. WHAT IS KNOWN ALREADY Embryos diagnosed as mosaic after PGT-A can develop into healthy infants, yet understanding the reasons behind their reproductive potential requires further research. One hypothesis suggests that mosaicism can be normalized through selective apoptosis and reduced proliferation of aneuploid cells, but direct evidence of these mechanisms in human embryos is lacking. Additionally, data interpretation from studies involving mosaic embryos has been hampered by retrospective analysis methods and the high incidence of false-positive mosaic diagnoses stemming from the use of poorly specific PGT-A platforms. STUDY DESIGN, SIZE, DURATION Prospective cohort study performing colocalization of cell-lineage and apoptotic markers by immunofluorescence (IF). We included a total of 64 human blastocysts donated to research on Day 5 or 6 post-fertilization (dpf) by 43 couples who underwent in vitro fertilization treatment with PGT-A at IVI-RMA Valencia between September 2019 and October 2022. A total of 27 mosaic blastocysts were analyzed. PARTICIPANTS/MATERIALS, SETTING, METHODS The study consisted of two phases: Phase I (caspase-3, n = 53 blastocysts): n = 13 euploid, n = 22 mosaic, n = 18 aneuploid. Phase II (terminal deoxynucleotidyl transferase dUTP nick end labelling (TUNEL), n = 11 blastocysts): n = 2 euploid, n = 5 mosaic, n = 4 aneuploid. Following donation for research, vitrified blastocysts were warmed, cultured until re-expansion, fixed, processed for IF, and imaged using confocal microscopy. For each blastocyst, the following cell counts were conducted: total cells (DAPI+), TE cells (GATA3+), inner cell mass (ICM) cells (GATA3-/NANOG+), and apoptotic cells (caspase-3+ or TUNEL+). The incidence of apoptosis was calculated for each blastocyst by dividing the number of caspase-3+ cells (Phase I) or TUNEL+ cells (Phase II) by the number of TE or ICM cells. Statistical analysis was performed according to data type and distribution (P < 0.05 was considered statistically significant). MAIN RESULTS AND THE ROLE OF CHANCE Phase I: Mosaic blastocysts displayed a similar number of total cells (49.6 ± 15 cells at 5 dpf; 58.8 ± 16.9 cells at 6 dpf), TE cells (38.8 ± 13.7 cells at 5 dpf; 49.2 ± 16.2 cells at 6 dpf), and ICM cells (10.9 ± 4.2 cells at 5 dpf; 9.7 ± 7.1 cells at 6 dpf) compared to euploid and aneuploid blastocysts (P > 0.05). The proportion of TE cells retaining NANOG expression increased gradually from euploid blastocysts (9.7% = 63/651 cells at 5 dpf; 0% = 0/157 cells at 6 dpf) to mosaic blastocysts (13.1% = 104/794 cells at 5 dpf; 3.4% = 12/353 cells at 6 dpf) and aneuploid blastocysts (27.9% = 149/534 cells at 5 dpf; 4.6% = 19/417 cells at 6 dpf) (P < 0.05). At the TE level, caspase-3+ cells were frequently observed (39% = 901/2310 cells). The proportion of caspase-3+ TE cells was significantly higher in mosaic blastocysts (44.1% ± 19.6 at 5 dpf; 43% ± 16.8 at 6 dpf) and aneuploid blastocysts (45.9% ± 16.1 at 5 dpf; 49% ± 15.1 at 6 dpf) compared to euploid blastocysts (26.6% ± 16.6 at 5 dpf; 17.5% ± 14.8 at 6 dpf) (P < 0.05). In contrast, at the ICM level, caspase-3+ cells were rarely observed (1.9% = 11/596 cells), and only detected in mosaic blastocysts (2.6% = 6/232 cells) and aneuploid blastocysts (2.5% = 5/197 cells) (P > 0.05). Phase II: Consistently, TUNEL+ cells were only observed in TE cells (32.4% = 124/383 cells). An increasing trend was identified toward a higher proportion of TUNEL+ cells in the TE of mosaic blastocysts (37.2% ± 21.9) and aneuploid blastocysts (39% ± 41.7), compared to euploid blastocysts (23% ± 32.5), although these differences did not reach statistical significance (P > 0.05). LIMITATIONS, REASONS FOR CAUTION The observed effects on apoptosis and differentiation may not be exclusive to aneuploid cells. Additionally, variations in aneuploidies and unexplored factors related to blastocyst development and karyotype concordance may introduce potential biases and uncertainties in the results. WIDER IMPLICATIONS OF THE FINDINGS Our findings demonstrate a cell lineage-specific effect of aneuploidy on the apoptotic levels and differentiation capacity of human blastocysts. This contributes to unravelling the biological characteristics of mosaic blastocysts and supports the concept of clonal depletion of aneuploid cells in explaining their reproductive potential. STUDY FUNDING/COMPETING INTEREST(S) This work was funded by grants from Centro para el Desarrollo Tecnológico Industrial (CDTI) (20190022) and Generalitat Valenciana (APOTIP/2019/009). None of the authors has any conflict of interest to declare. TRIAL REGISTRATION NUMBER N/A.
Collapse
Affiliation(s)
- Ángel Martín
- Department of Innovation, IVIRMA Global Research Alliance, IVI Foundation, Health Research Institute La Fe, Valencia, Spain
| | - Amparo Mercader
- Department of Innovation, IVIRMA Global Research Alliance, IVI Foundation, Health Research Institute La Fe, Valencia, Spain
- Department of Research, IVF Laboratory, IVIRMA Global, Valencia, Spain
| | - Diana Beltrán
- Department of Research, IVF Laboratory, IVIRMA Global, Valencia, Spain
| | - Amparo Mifsud
- Department of Research, IVF Laboratory, IVIRMA Global, Valencia, Spain
| | - Mar Nohales
- Department of Research, IVF Laboratory, IVIRMA Global, Valencia, Spain
| | - María Luisa Pardiñas
- Department of Innovation, IVIRMA Global Research Alliance, IVI Foundation, Health Research Institute La Fe, Valencia, Spain
| | - David Ortega-Jaén
- Department of Innovation, IVIRMA Global Research Alliance, IVI Foundation, Health Research Institute La Fe, Valencia, Spain
| | - María José de Los Santos
- Department of Innovation, IVIRMA Global Research Alliance, IVI Foundation, Health Research Institute La Fe, Valencia, Spain
- Department of Research, IVF Laboratory, IVIRMA Global, Valencia, Spain
| |
Collapse
|
23
|
Robertson SA, Richards RI. Single-cell sequencing shows mosaic aneuploidy in most human embryos. J Clin Invest 2024; 134:e179134. [PMID: 38488008 PMCID: PMC10940079 DOI: 10.1172/jci179134] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/18/2024] Open
Abstract
Mammalian preimplantation embryos often contain chromosomal defects that arose in the first divisions after fertilization and affect a subpopulation of cells - an event known as mosaic aneuploidy. In this issue of the JCI, Chavli et al. report single-cell genomic sequencing data for rigorous evaluation of the incidence and degree of mosaic aneuploidy in healthy human in vitro fertilization (IVF) embryos. Remarkably, mosaic aneuploidy occurred in at least 80% of human blastocyst-stage embryos, with often less than 20% of cells showing defects. These findings confirm that mosaic aneuploidy is prevalent in human embryos, indicating that the process is a widespread event that rarely has clinical consequences. There are major implications for preimplantation genetic testing of aneuploidy (PGT-A), a test commonly used to screen and select IVF embryos for transfer. The application and benefit of this technology is controversial, and the findings provide more cause for caution on its use.
Collapse
Affiliation(s)
| | - Robert I. Richards
- School of Molecular Bioscience, University of Adelaide, Adelaide, South Australia, Australia
| |
Collapse
|
24
|
Fischer F, Ernst L, Frey A, Holstein K, Prasad D, Weichselberger V, Balaji R, Classen AK. A mismatch in the expression of cell surface molecules induces tissue-intrinsic defense against aberrant cells. Curr Biol 2024; 34:980-996.e6. [PMID: 38350446 DOI: 10.1016/j.cub.2024.01.053] [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: 03/09/2023] [Revised: 09/29/2023] [Accepted: 01/19/2024] [Indexed: 02/15/2024]
Abstract
Tissue-intrinsic error correction enables epithelial cells to detect abnormal neighboring cells and facilitate their removal from the tissue. One of these pathways, "interface surveillance," is triggered by cells with aberrant developmental and cell-fate-patterning pathways. It remains unknown which molecular mechanisms provide cells with the ability to compare fate between neighboring cells. We demonstrate that Drosophila imaginal discs express an array of cell surface molecules previously implicated in neuronal axon guidance processes. They include members of the Robo, Teneurin, Ephrin, Toll-like, or atypical cadherin families. Importantly, a mismatch in expression levels of these cell surface molecules between adjacent cells is sufficient to induce interface surveillance, indicating that differences in expression levels between neighboring cells, rather than their absolute expression levels, are crucial. Specifically, a mismatch in Robo2 and Robo3, but not Robo1, induces enrichment of actin, myosin II, and Ena/Vasp, as well as activation of JNK and apoptosis at clonal interfaces. Moreover, Robo2 can induce interface surveillance independently of its cytosolic domain and without the need for the Robo-ligand Slit. The expression of Robo2 and other cell surface molecules, such as Teneurins or the Ephrin receptor is regulated by fate-patterning pathways intrinsic and extrinsic to the wing disc, as well as by expression of oncogenic RasV12. Combined, we demonstrate that neighboring cells respond to a mismatch in surface code patterns mediated by specific transmembrane proteins and reveal a novel function for these cell surface proteins in cell fate recognition and removal of aberrant cells during development and homeostasis of epithelial tissues.
Collapse
Affiliation(s)
- Friedericke Fischer
- Hilde-Mangold-Haus, University of Freiburg, 79104 Freiburg, Germany; Faculty of Biology, University of Freiburg, 79104 Freiburg, Germany; International Max Planck Research School for Immunobiology, Epigenetics, and Metabolism, 79108 Freiburg, Germany
| | - Laurin Ernst
- Hilde-Mangold-Haus, University of Freiburg, 79104 Freiburg, Germany; Faculty of Biology, University of Freiburg, 79104 Freiburg, Germany; International Max Planck Research School for Immunobiology, Epigenetics, and Metabolism, 79108 Freiburg, Germany
| | - Anna Frey
- Hilde-Mangold-Haus, University of Freiburg, 79104 Freiburg, Germany; Faculty of Biology, University of Freiburg, 79104 Freiburg, Germany; Spemann Graduate School of Biology and Medicine (SGBM), University of Freiburg, 79104 Freiburg, Germany
| | - Katrin Holstein
- Department of Vascular Cell Biology, Max Planck Institute for Molecular Biomedicine, 48149 Münster, Germany
| | - Deepti Prasad
- Hilde-Mangold-Haus, University of Freiburg, 79104 Freiburg, Germany; Faculty of Biology, University of Freiburg, 79104 Freiburg, Germany; Spemann Graduate School of Biology and Medicine (SGBM), University of Freiburg, 79104 Freiburg, Germany
| | - Vanessa Weichselberger
- Spemann Graduate School of Biology and Medicine (SGBM), University of Freiburg, 79104 Freiburg, Germany; Aix Marseille University, CNRS, UMR 7288, IBDM, 13288 Marseille, France
| | - Ramya Balaji
- Hilde-Mangold-Haus, University of Freiburg, 79104 Freiburg, Germany; Faculty of Biology, University of Freiburg, 79104 Freiburg, Germany
| | - Anne-Kathrin Classen
- Hilde-Mangold-Haus, University of Freiburg, 79104 Freiburg, Germany; Faculty of Biology, University of Freiburg, 79104 Freiburg, Germany; CIBSS Centre for Integrative Biological Signalling Studies, University of Freiburg, 79104 Freiburg, Germany; BIOSS Centre for Biological Signalling Studies, University of Freiburg, 79104 Freiburg, Germany.
| |
Collapse
|
25
|
Budrewicz J, Chavez SL. Insights into embryonic chromosomal instability: mechanisms of DNA elimination during mammalian preimplantation development. Front Cell Dev Biol 2024; 12:1344092. [PMID: 38374891 PMCID: PMC10875028 DOI: 10.3389/fcell.2024.1344092] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2023] [Accepted: 01/15/2024] [Indexed: 02/21/2024] Open
Abstract
Mammalian preimplantation embryos often contend with aneuploidy that arose either by the inheritance of meiotic errors from the gametes, or from mitotic mis-segregation events that occurred following fertilization. Regardless of the origin, mis-segregated chromosomes become encapsulated in micronuclei (MN) that are spatially isolated from the main nucleus. Much of our knowledge of MN formation comes from dividing somatic cells during tumorigenesis, but the error-prone cleavage-stage of early embryogenesis is fundamentally different. One unique aspect is that cellular fragmentation (CF), whereby small subcellular bodies pinch off embryonic blastomeres, is frequently observed. CF has been detected in both in vitro and in vivo-derived embryos and likely represents a response to chromosome mis-segregation since it only appears after MN formation. There are multiple fates for MN, including sequestration into CFs, but the molecular mechanism(s) by which this occurs remains unclear. Due to nuclear envelope rupture, the chromosomal material contained within MN and CFs becomes susceptible to double stranded-DNA breaks. Despite this damage, embryos may still progress to the blastocyst stage and exclude chromosome-containing CFs, as well as non-dividing aneuploid blastomeres, from participating in further development. Whether these are attempts to rectify MN formation or eliminate embryos with poor implantation potential is unknown and this review will discuss the potential implications of DNA removal by CF/blastomere exclusion. We will also extrapolate what is known about the intracellular pathways mediating MN formation and rupture in somatic cells to preimplantation embryogenesis and how nuclear budding and DNA release into the cytoplasm may impact overall development.
Collapse
Affiliation(s)
- Jacqueline Budrewicz
- Department of Molecular and Medical Genetics, Oregon Health and Science University, Portland, OR, United States
- Division of Reproductive and Developmental Sciences, Oregon National Primate Research Center, Beaverton, OR, United States
| | - Shawn L. Chavez
- Department of Molecular and Medical Genetics, Oregon Health and Science University, Portland, OR, United States
- Division of Reproductive and Developmental Sciences, Oregon National Primate Research Center, Beaverton, OR, United States
- Department of Obstetrics and Gynecology, Oregon Health and Science University, Portland, OR, United States
- Department of Biomedical Engineering, Oregon Health and Science University, Portland, OR, United States
| |
Collapse
|
26
|
Zou Y, Sui Y, Fu J, Ge N, Sun X, Sun Y. The morphokinetic signature of human blastocysts with mosaicism and the clinical outcomes following transfer of embryos with low-level mosaicism. J Ovarian Res 2024; 17:10. [PMID: 38195558 PMCID: PMC10775590 DOI: 10.1186/s13048-023-01324-w] [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: 09/22/2023] [Accepted: 12/12/2023] [Indexed: 01/11/2024] Open
Abstract
BACKGROUND Genetic mosaicism is commonly observed in human blastocysts. Embryos' morphokinetic feature observed from time-lapse monitoring (TLM) is helpful to predict the embryos' ploidy status in a non-invasive way. However, morphokinetic research on mosaic embryos is extremely limited. Moreover, transfer of mosaic embryos is a new attempt in reproductive medicine, while studies regarding the clinical and neonatal outcomes following transfer of embryos with different levels and types of mosaicism are needed. This study aimed to investigate the morphokinetic characteristics of mosaic blastocysts, uncover clinical outcomes of mosaic embryos, and evaluate the effect of level and type of mosaicism on transfer outcomes. RESULTS A total of 923 blastocysts from 229 preimplantation genetic testing cycles were cultured in TLM incubators in a single fertilization center between July 2016 and July 2021. Multivariate logistic regression models showed mosaic embryos had significantly shorter time to reach morula when compared with euploid (P = 0.002), mosaic with aneuploid (P = 0.005), and aneuploid (P = 0.005) embryos after adjusting the potential confounders. KIDScore is an artificial intelligence scoring program from time lapse incubation system to predict embryo implantation potential. Mosaic with aneuploid embryos had significantly lower KIDScore than euploid (P = 6.47e-4), mosaic (P = 0.005), and aneuploid (P = 0.004) embryos after adjustment. Meanwhile, we compared the clinical outcomes following transfer of low-level (< 50%) mosaic embryos (N = 60) with euploid embryos (N = 1301) matched using propensity scoring collected from September 2020 to January 2023. Mosaic embryos had significantly lower clinical pregnancy rate (41.67% vs. 57.65%, P = 0.015) and live birth rate (38.33% vs. 51.35%, P = 0.048) than the euploid embryos. Subgroup analyses showed the whole, segmental, and complex chromosome mosaic embryos had the similar clinical outcomes. CONCLUSIONS The shortened time to reach morula in mosaic embryos and the low KIDScore in mosaic with aneuploid embryos revealed innovative clues to embryo selection with the non-invasive TLM and provided new insights into biological mechanism of chromosomal abnormality. The analyses of overall and subgroups of mosaic embryo transfer outcomes helped to optimize embryo transfer scheme for in-vitro fertilization procedures. Multi-center prospective studies with large sample sizes are warranted to validate our results in the future.
Collapse
Affiliation(s)
- Yaoyu Zou
- Shanghai Ji Ai Genetics & IVF Institute, Obstetrics & Gynecology Hospital, Fudan University, Dalin Road, Shanghai, 200011, China
| | - Yilun Sui
- Shanghai Ji Ai Genetics & IVF Institute, Obstetrics & Gynecology Hospital, Fudan University, Dalin Road, Shanghai, 200011, China
| | - Jing Fu
- Shanghai Ji Ai Genetics & IVF Institute, Obstetrics & Gynecology Hospital, Fudan University, Dalin Road, Shanghai, 200011, China
| | - Naidong Ge
- Shanghai Ji Ai Genetics & IVF Institute, Obstetrics & Gynecology Hospital, Fudan University, Dalin Road, Shanghai, 200011, China
| | - Xiaoxi Sun
- Shanghai Ji Ai Genetics & IVF Institute, Obstetrics & Gynecology Hospital, Fudan University, Dalin Road, Shanghai, 200011, China.
| | - Yijuan Sun
- Shanghai Ji Ai Genetics & IVF Institute, Obstetrics & Gynecology Hospital, Fudan University, Dalin Road, Shanghai, 200011, China.
| |
Collapse
|
27
|
Chavli EA, Klaasen SJ, Van Opstal D, Laven JS, Kops GJ, Baart EB. Single-cell DNA sequencing reveals a high incidence of chromosomal abnormalities in human blastocysts. J Clin Invest 2024; 134:e174483. [PMID: 38175717 PMCID: PMC10940095 DOI: 10.1172/jci174483] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Accepted: 01/02/2024] [Indexed: 01/05/2024] Open
Abstract
Aneuploidy, a deviation from the normal chromosome copy number, is common in human embryos and is considered a primary cause of implantation failure and early pregnancy loss. Meiotic errors lead to uniformly abnormal karyotypes, while mitotic errors lead to chromosomal mosaicism: the presence of cells with at least 2 different karyotypes within an embryo. Knowledge about mosaicism in blastocysts mainly derives from bulk DNA sequencing (DNA-Seq) of multicellular trophectoderm (TE) and/or inner cell mass (ICM) samples. However, this can only detect an average net gain or loss of DNA above a detection threshold of 20%-30%. To accurately assess mosaicism, we separated the TE and ICM of 55 good-quality surplus blastocysts and successfully applied single-cell whole-genome sequencing (scKaryo-Seq) on 1,057 cells. Mosaicism involving numerical and structural chromosome abnormalities was detected in 82% of the embryos, in which most abnormalities affected less than 20% of the cells. Structural abnormalities, potentially caused by replication stress and DNA damage, were observed in 69% of the embryos. In conclusion, our findings indicated that mosaicism was prevalent in good-quality blastocysts, whereas these blastocysts would likely be identified as normal with current bulk DNA-Seq techniques used for preimplantation genetic testing for aneuploidy.
Collapse
Affiliation(s)
- Effrosyni A. Chavli
- Division of Reproductive Endocrinology and Infertility, Department of Obstetrics and Gynecology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, Netherlands
| | - Sjoerd J. Klaasen
- Hubrecht Institute-KNAW (Royal Academy of Arts and Sciences) and University Medical Center Utrecht, Utrecht, Netherlands
- Oncode Institute, Utrecht, Netherlands
| | | | - Joop S.E. Laven
- Division of Reproductive Endocrinology and Infertility, Department of Obstetrics and Gynecology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, Netherlands
| | - Geert J.P.L. Kops
- Hubrecht Institute-KNAW (Royal Academy of Arts and Sciences) and University Medical Center Utrecht, Utrecht, Netherlands
- Oncode Institute, Utrecht, Netherlands
| | - Esther B. Baart
- Division of Reproductive Endocrinology and Infertility, Department of Obstetrics and Gynecology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, Netherlands
- Department of Developmental Biology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, Netherlands
| |
Collapse
|
28
|
Campos G, Sciorio R, Fleming S. Healthy Live Births after the Transfer of Mosaic Embryos: Self-Correction or PGT-A Overestimation? Genes (Basel) 2023; 15:18. [PMID: 38275600 PMCID: PMC10815078 DOI: 10.3390/genes15010018] [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: 12/04/2023] [Revised: 12/18/2023] [Accepted: 12/20/2023] [Indexed: 01/27/2024] Open
Abstract
The implementation of next generation sequencing (NGS) in preimplantation genetic testing for aneuploidy (PGT-A) has led to a higher prevalence of mosaic diagnosis within the trophectoderm (TE) sample. Regardless, mosaicism could potentially increase the rate of live-born children with chromosomic syndromes, though available data from the transfer of embryos with putative PGT-A mosaicism are scarce but reassuring. Even with lower implantation and higher miscarriage rates, mosaic embryos can develop into healthy live births. Therefore, this urges an explanation for the disappearance of aneuploid cells throughout development, to provide guidance in the management of mosaicism in clinical practice. Technical overestimation of mosaicism, together with some sort of "self-correction" mechanisms during the early post-implantation stages, emerged as potential explanations. Unlike the animal model, in which the elimination of genetically abnormal cells from the future fetal lineage has been demonstrated, in human embryos this capability remains unverified even though the germ layer displays an aneuploidy-induced cell death lineage preference with higher rates of apoptosis in the inner cell mass (ICM) than in the TE cells. Moreover, the reported differential dynamics of cell proliferation and apoptosis between euploid, mosaic, and aneuploid embryos, together with pro-apoptosis gene products (cfDNA and mRNA) and extracellular vesicles identified in the blastocoel fluid, may support the hypothesis of apoptosis as a mechanism to purge the preimplantation embryo of aneuploid cells. Alternative hypotheses, like correction of aneuploidy by extrusion of a trisomy chromosome or by monosomic chromosome duplication, are even, though they represent an extremely rare phenomenon. On the other hand, the technical limitations of PGT-A analysis may lead to inaccuracy in embryo diagnoses, identifying as "mosaic" those embryos that are uniformly euploid or aneuploid. NGS assumption of "intermediate copy number profiles" as evidence of a mixture of euploid and aneuploid cells in a single biopsy has been reported to be poorly predictive in cases of mosaicism diagnosis. Additionally, the concordance found between the TE and the ICM in cases of TE biopsies displaying mosaicism is lower than expected, and it correlates differently depending on the type (whole chromosome versus segmental) and the level of mosaicism reported. Thus, in cases of low-/medium-level mosaicism (<50%), aneuploid cells would rarely involve the ICM and other regions. However, in high-level mosaics (≥50%), abnormal cells in the ICM should display higher prevalence, revealing more uniform aneuploidy in most embryos, representing a technical variation in the uniform aneuploidy range, and therefore might impair the live birth rate.
Collapse
Affiliation(s)
- Gerard Campos
- Geisinger Medical Center, Women’s Health Fertility Clinic, Danville, PA 17821, USA;
- GIREXX Fertility Clinics, C. de Cartagena, 258, 08025 Girona, Spain
| | - Romualdo Sciorio
- Fertility Medicine and Gynaecological Endocrinology Unit, Department Woman-Mother-Child, Lausanne University Hospital, 1011 Lausanne, Switzerland
| | - Steven Fleming
- Discipline of Anatomy & Histology, School of Medical Sciences, University of Sydney, Sydney, NSW 2006, Australia;
| |
Collapse
|
29
|
Essers R, Lebedev IN, Kurg A, Fonova EA, Stevens SJC, Koeck RM, von Rango U, Brandts L, Deligiannis SP, Nikitina TV, Sazhenova EA, Tolmacheva EN, Kashevarova AA, Fedotov DA, Demeneva VV, Zhigalina DI, Drozdov GV, Al-Nasiry S, Macville MVE, van den Wijngaard A, Dreesen J, Paulussen A, Hoischen A, Brunner HG, Salumets A, Zamani Esteki M. Prevalence of chromosomal alterations in first-trimester spontaneous pregnancy loss. Nat Med 2023; 29:3233-3242. [PMID: 37996709 PMCID: PMC10719097 DOI: 10.1038/s41591-023-02645-5] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Accepted: 10/11/2023] [Indexed: 11/25/2023]
Abstract
Pregnancy loss is often caused by chromosomal abnormalities of the conceptus. The prevalence of these abnormalities and the allocation of (ab)normal cells in embryonic and placental lineages during intrauterine development remain elusive. In this study, we analyzed 1,745 spontaneous pregnancy losses and found that roughly half (50.4%) of the products of conception (POCs) were karyotypically abnormal, with maternal and paternal age independently contributing to the increased genomic aberration rate. We applied genome haplarithmisis to a subset of 94 pregnancy losses with normal parental and POC karyotypes. Genotyping of parental DNA as well as POC extra-embryonic mesoderm and chorionic villi DNA, representing embryonic and trophoblastic tissues, enabled characterization of the genomic landscape of both lineages. Of these pregnancy losses, 35.1% had chromosomal aberrations not previously detected by karyotyping, increasing the rate of aberrations of pregnancy losses to 67.8% by extrapolation. In contrast to viable pregnancies where mosaic chromosomal abnormalities are often restricted to chorionic villi, such as confined placental mosaicism, we found a higher degree of mosaic chromosomal imbalances in extra-embryonic mesoderm rather than chorionic villi. Our results stress the importance of scrutinizing the full allelic architecture of genomic abnormalities in pregnancy loss to improve clinical management and basic research of this devastating condition.
Collapse
Affiliation(s)
- Rick Essers
- Department of Clinical Genetics, Maastricht University Medical Center (MUMC+), Maastricht, The Netherlands
- Department of Genetics and Cell Biology, GROW-Research Institute for Oncology and Reproduction, Faculty of Health, Medicine and Life Sciences (FHML), Maastricht University, Maastricht, The Netherlands
| | - Igor N Lebedev
- Research Institute of Medical Genetics, Tomsk National Research Medical Center of the Russian Academy of Sciences, Tomsk, Russia
| | - Ants Kurg
- Department of Biotechnology, Institute of Molecular and Cell Biology, University of Tartu, Tartu, Estonia
| | - Elizaveta A Fonova
- Research Institute of Medical Genetics, Tomsk National Research Medical Center of the Russian Academy of Sciences, Tomsk, Russia
| | - Servi J C Stevens
- Department of Clinical Genetics, Maastricht University Medical Center (MUMC+), Maastricht, The Netherlands
- Department of Genetics and Cell Biology, GROW-Research Institute for Oncology and Reproduction, Faculty of Health, Medicine and Life Sciences (FHML), Maastricht University, Maastricht, The Netherlands
| | - Rebekka M Koeck
- Department of Clinical Genetics, Maastricht University Medical Center (MUMC+), Maastricht, The Netherlands
- Department of Genetics and Cell Biology, GROW-Research Institute for Oncology and Reproduction, Faculty of Health, Medicine and Life Sciences (FHML), Maastricht University, Maastricht, The Netherlands
| | - Ulrike von Rango
- Department of Anatomy & Embryology, Faculty of Health, Medicine and Life Sciences (FHML), Maastricht University, Maastricht, The Netherlands
| | - Lloyd Brandts
- Department of Clinical Epidemiology and Medical Technology Assessment (KEMTA), Maastricht University Medical Center (MUMC+), Maastricht, The Netherlands
| | - Spyridon Panagiotis Deligiannis
- Department of Obstetrics and Gynaecology, Institute of Clinical Medicine, University of Tartu, Tartu, Estonia
- Department of Obstetrics and Gynecology, University of Helsinki, Helsinki, Finland
| | - Tatyana V Nikitina
- Research Institute of Medical Genetics, Tomsk National Research Medical Center of the Russian Academy of Sciences, Tomsk, Russia
| | - Elena A Sazhenova
- Research Institute of Medical Genetics, Tomsk National Research Medical Center of the Russian Academy of Sciences, Tomsk, Russia
| | - Ekaterina N Tolmacheva
- Research Institute of Medical Genetics, Tomsk National Research Medical Center of the Russian Academy of Sciences, Tomsk, Russia
| | - Anna A Kashevarova
- Research Institute of Medical Genetics, Tomsk National Research Medical Center of the Russian Academy of Sciences, Tomsk, Russia
| | - Dmitry A Fedotov
- Research Institute of Medical Genetics, Tomsk National Research Medical Center of the Russian Academy of Sciences, Tomsk, Russia
| | - Viktoria V Demeneva
- Research Institute of Medical Genetics, Tomsk National Research Medical Center of the Russian Academy of Sciences, Tomsk, Russia
| | - Daria I Zhigalina
- Research Institute of Medical Genetics, Tomsk National Research Medical Center of the Russian Academy of Sciences, Tomsk, Russia
| | - Gleb V Drozdov
- Research Institute of Medical Genetics, Tomsk National Research Medical Center of the Russian Academy of Sciences, Tomsk, Russia
| | - Salwan Al-Nasiry
- Department of Genetics and Cell Biology, GROW-Research Institute for Oncology and Reproduction, Faculty of Health, Medicine and Life Sciences (FHML), Maastricht University, Maastricht, The Netherlands
- Department of Obstetrics and Gynaecology, Maastricht University Medical Center (MUMC+), Maastricht, The Netherlands
| | - Merryn V E Macville
- Department of Clinical Genetics, Maastricht University Medical Center (MUMC+), Maastricht, The Netherlands
- Department of Genetics and Cell Biology, GROW-Research Institute for Oncology and Reproduction, Faculty of Health, Medicine and Life Sciences (FHML), Maastricht University, Maastricht, The Netherlands
| | - Arthur van den Wijngaard
- Department of Clinical Genetics, Maastricht University Medical Center (MUMC+), Maastricht, The Netherlands
- Department of Genetics and Cell Biology, GROW-Research Institute for Oncology and Reproduction, Faculty of Health, Medicine and Life Sciences (FHML), Maastricht University, Maastricht, The Netherlands
| | - Jos Dreesen
- Department of Clinical Genetics, Maastricht University Medical Center (MUMC+), Maastricht, The Netherlands
| | - Aimee Paulussen
- Department of Clinical Genetics, Maastricht University Medical Center (MUMC+), Maastricht, The Netherlands
- Department of Genetics and Cell Biology, GROW-Research Institute for Oncology and Reproduction, Faculty of Health, Medicine and Life Sciences (FHML), Maastricht University, Maastricht, The Netherlands
| | - Alexander Hoischen
- Department of Human Genetics, Radboud University Medical Center, Nijmegen, The Netherlands
- Department of Internal Medicine, Center for Infectious Disease (RCI), Radboud University Medical Center, Nijmegen, The Netherlands
- Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
- Radboud Expertise Center for Immunodeficiency and Autoinflammation and Radboud Center for Infectious Disease (RCI), Radboud University Medical Center, Nijmegen, The Netherlands
| | - Han G Brunner
- Department of Clinical Genetics, Maastricht University Medical Center (MUMC+), Maastricht, The Netherlands
- Department of Genetics and Cell Biology, GROW-Research Institute for Oncology and Reproduction, Faculty of Health, Medicine and Life Sciences (FHML), Maastricht University, Maastricht, The Netherlands
- Department of Human Genetics, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Andres Salumets
- Department of Obstetrics and Gynaecology, Institute of Clinical Medicine, University of Tartu, Tartu, Estonia.
- Competence Center on Health Technologies, Tartu, Estonia.
- Division of Obstetrics and Gynecology, Department of Clinical Science, Intervention & Technology (CLINTEC), Karolinska Institutet and Karolinska University Hospital, Stockholm, Sweden.
| | - Masoud Zamani Esteki
- Department of Clinical Genetics, Maastricht University Medical Center (MUMC+), Maastricht, The Netherlands.
- Department of Genetics and Cell Biology, GROW-Research Institute for Oncology and Reproduction, Faculty of Health, Medicine and Life Sciences (FHML), Maastricht University, Maastricht, The Netherlands.
- Division of Obstetrics and Gynecology, Department of Clinical Science, Intervention & Technology (CLINTEC), Karolinska Institutet and Karolinska University Hospital, Stockholm, Sweden.
| |
Collapse
|
30
|
Campos G. PGT-A mosaicism based on NGS intermediate copy numbers: is it time to stop reporting them? J Assist Reprod Genet 2023; 40:2925-2932. [PMID: 37735311 PMCID: PMC10656405 DOI: 10.1007/s10815-023-02936-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Accepted: 09/06/2023] [Indexed: 09/23/2023] Open
Abstract
Mosaicism represents a genuine real phenomenon, but its high prevalence and undisclosed clinical significance, stress the burden on genetic counseling and the management of PGT-A results. Even though the assumption of mosaicism from NGS intermediate chromosome copy number profiles may represent a reasonable interpretation, other potential technical reasons, including amplification bias, contamination, biopsy technique, or the analysis algorithms, may constitute alternative explanations. Thresholds confining mosaicism ranges are established according to models employing mixtures of normal and abnormal cells with steady conditions of quantity and quality which are unable to reflect the full extent of variability present in a trophectoderm (TE) biopsy specimen. When the concordance of TE with the ICM is considered, mosaic TE biopsies poorly correlate with the chromosomal status of the remaining embryo, displaying mostly ICM aneuploidy in cases of TE high-range mosaics diagnosis and euploidy when mosaicism grade in TE is less than 50% (low-mid range mosaicism), which implies an evident overestimation of mosaicism results. Indeed, a binary classification of NGS profiles that excludes mosaic ranges, including only euploid and aneuploid diagnosis, provides higher specificity and accuracy in identifying abnormal embryos and discarding them. As intermediate copy number profiles do not represent strong evidence of mosaicism but only an inaccurate and misleading assumption, and considering that no increased risk has been reported in the offspring, until diagnosis specificity is improved and its clinical implications are determined, laboratories should consider limiting predictions to euploid and aneuploid and stop reporting mosaicism.
Collapse
Affiliation(s)
- Gerard Campos
- Geisinger Medical Center, Women's Health Fertility Clinic, Danville, PA, USA.
- GIREXX Fertility Clinics, Barcelona, Spain.
| |
Collapse
|
31
|
Zhong W, Shen K, Xue X, Wang W, Wang W, Zuo H, Guo Y, Yao S, Sun M, Song C, Wang Q, Ruan Z, Yao X, Shang W. Single-cell multi-omics sequencing reveals chromosome copy number inconsistency between trophectoderm and inner cell mass in human reconstituted embryos after spindle transfer. Hum Reprod 2023; 38:2137-2153. [PMID: 37766497 DOI: 10.1093/humrep/dead186] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Revised: 08/06/2023] [Indexed: 09/29/2023] Open
Abstract
STUDY QUESTION Is the chromosome copy number of the trophectoderm (TE) of a human reconstituted embryos after spindle transfer (ST) representative of the inner cell mass (ICM)? SUMMARY ANSWER Single-cell multi-omics sequencing revealed that ST blastocysts have a higher proportion of cell lineages exhibiting intermediate mosaicism than conventional ICSI blastocysts, and that the TE of ST blastocysts does not represent the chromosome copy number of ICM. WHAT IS KNOWN ALREADY Preimplantation genetic testing for aneuploidy (PGT-A) assumes that TE biopsies are representative of the ICM, but the TE and ICM originate from different cell lineages, and concordance between TE and ICM is not well-studied, especially in ST embryos. STUDY DESIGN, SIZE, DURATION We recruited 30 infertile women who received treatment at our clinic and obtained 45 usable blastocysts (22 from conventional ICSI and 23 reconstituted embryos after ST). We performed single-cell multi-omics sequencing on all blastocysts to predict and verify copy number variations (CNVs) in each cell. We determined the chromosome copy number of each embryo by analysing the proportion of abnormal cells in each blastocyst. We used the Bland-Altman concordance and the Kappa test to evaluate the concordance between TE and ICM in the both groups. PARTICIPANTS/MATERIALS, SETTING, METHODS The study was conducted at a public tertiary hospital in China, where all the embryo operations, including oocytes retrieval, ST, and ICSI, were performed in the embryo laboratory. We utilized single-cell multi-omics sequencing technology at the Biomedical Pioneering Innovation Center, School of Life Sciences, Peking University, to analyse the blastocysts. Transcriptome sequencing was used to predict the CNV of each cell through bioinformatics analysis, and the results were validated using the DNA methylation library of each cell to confirm chromosomal normalcy. We conducted statistical analysis and graphical plotting using R 4.2.1, SPSS 27, and GraphPad Prism 9.3. MAIN RESULTS AND THE ROLE OF CHANCE Mean age of the volunteers, the blastocyst morphology, and the developmental ratewere similar in ST and ICSI groups. The blastocysts in the ST group had some additional chromosomal types that were prone to variations beyond those enriched in the blastocysts of the ICSI group. Finally, both Bland-Altman concordance test and kappa concordancetest showed good chromosomal concordance between TE and ICM in the ICSI blastocysts (kappa = 0.659, P < 0.05), but not in ST blastocysts (P = 1.000), suggesting that the TE in reconstituted embryos is not representative of ICM. Gene functional annotation (GO and KEGG analyses) suggests that there may be new or additional pathways for CNV generation in ST embryos compared to ICSI embryos. LIMITATIONS, REASONS FOR CAUTION This study was mainly limited by the small sample size and the limitations of single-cell multi-omics sequencing technology. To select eligible single cells, some cells of the embryos were eliminated or not labelled, resulting in a loss of information about them. The findings of this study are innovative and exploratory. A larger sample size of human embryos (especially ST embryos) and more accurate molecular genetics techniques for detecting CNV in single cells are needed to validate our results. WIDER IMPLICATIONS OF THE FINDINGS Our study justifies the routine clinical use of PGT-A in ICSI blastocysts, as we found that the TE is a good substitute for ICM in predicting chromosomal abnormalities. While PGT-A is not entirely accurate, our data demonstrate good clinical feasibility. This trial was able to provide correct genetic counselling to patients regarding the reliability of PGT-A. Regarding ST blastocysts, the increased mosaicism rate and the inability of the TE to represent the chromosomal copy number of the ICM are both biological characteristics that differentiate them from ICSI blastocysts. Currently, ST is not used clinically on a large scale to produce blastocysts. However, if ST becomes more widely used in the future, our study will be the first to demonstrate that the use of PGT-A in ST blastocysts may not be as accurate as PGT-A for ICSI blastocysts. STUDY FUNDING/COMPETING INTEREST(S) This study was supported by grants from the National Key R&D Program of China (2018YFA0107601) and the National Key R&D Program of China (2018YFC1003003). The authors declare no conflict of interest. TRIAL REGISTRATION NUMBER N/A.
Collapse
Affiliation(s)
- Wei Zhong
- School of Medicine, South China University of Technology, Guangzhou, China
- Department of Obstetrics and Gynecology, The Sixth Medical Center of PLA General Hospital of Beijing, Beijing, China
| | - Kexin Shen
- School of Medicine, South China University of Technology, Guangzhou, China
- Department of Obstetrics and Gynecology, The Sixth Medical Center of PLA General Hospital of Beijing, Beijing, China
| | - Xiaohui Xue
- Peking University-Tsinghua University-National Institute of Biological Sciences Joint Graduate Program, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, China
| | - Wei Wang
- Department of Obstetrics and Gynecology, The Seventh Medical Center of Chinese PLA General Hospital, Beijing, China
- Department of Obstetrics and Gynecology, Chinese PLA General Hospital, Beijing, China
| | - Weizhou Wang
- Department of Obstetrics and Gynecology, The Sixth Medical Center of PLA General Hospital of Beijing, Beijing, China
| | - Haiyang Zuo
- Department of Obstetrics and Gynecology, The Sixth Medical Center of PLA General Hospital of Beijing, Beijing, China
| | - Yiming Guo
- Department of Biological Science, Dietrich School Of Art and Science, University of Pittsburgh, Pittsburgh, PA, USA
| | - Shun Yao
- Department of Obstetrics and Gynecology, The Sixth Medical Center of PLA General Hospital of Beijing, Beijing, China
- Navy Clinical Medical School, Anhui Medical University, Hefei, China
| | - Mingyue Sun
- Department of Obstetrics and Gynecology, The Sixth Medical Center of PLA General Hospital of Beijing, Beijing, China
- Department of Histology and Embryology, Hebei Medical University, Shijiazhuang, Hebei, China
| | - Chunlan Song
- Department of Obstetrics and Gynecology, The Sixth Medical Center of PLA General Hospital of Beijing, Beijing, China
- Department of Obstetrics and Gynecology, Chinese PLA General Hospital, Beijing, China
| | - Qihang Wang
- Department of Obstetrics and Gynecology, The Seventh Medical Center of Chinese PLA General Hospital, Beijing, China
- Department of Obstetrics and Gynecology, Chinese PLA General Hospital, Beijing, China
| | - Zhuolin Ruan
- Department of Obstetrics and Gynecology, Chinese PLA General Hospital, Beijing, China
| | - Xinyi Yao
- Department of Obstetrics and Gynecology, Chinese PLA General Hospital, Beijing, China
| | - Wei Shang
- School of Medicine, South China University of Technology, Guangzhou, China
- Department of Obstetrics and Gynecology, The Sixth Medical Center of PLA General Hospital of Beijing, Beijing, China
- Department of Obstetrics and Gynecology, The Seventh Medical Center of Chinese PLA General Hospital, Beijing, China
- Department of Obstetrics and Gynecology, Chinese PLA General Hospital, Beijing, China
| |
Collapse
|
32
|
Viotti M, Greco E, Grifo JA, Madjunkov M, Librach C, Cetinkaya M, Kahraman S, Yakovlev P, Kornilov N, Corti L, Biricik A, Cheng EH, Su CY, Lee MS, Bonifacio MD, Cooper AR, Griffin DK, Tran DY, Kaur P, Barnes FL, Zouves CG, Victor AR, Besser AG, Madjunkova S, Spinella F. Chromosomal, gestational, and neonatal outcomes of embryos classified as a mosaic by preimplantation genetic testing for aneuploidy. Fertil Steril 2023; 120:957-966. [PMID: 37532168 DOI: 10.1016/j.fertnstert.2023.07.022] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2023] [Revised: 07/25/2023] [Accepted: 07/25/2023] [Indexed: 08/04/2023]
Abstract
OBJECTIVE To understand the clinical risks associated with the transfer of embryos classified as a mosaic using preimplantation genetic testing for aneuploidy. DESIGN Analysis of data collected between 2017 and 2023. SETTING Multicenter. PATIENTS Patients of infertility treatment. INTERVENTION Comparison of pregnancies resulting from embryos classified as euploid or mosaic using the 20%-80% interval in chromosomal intermediate copy numbers to define a mosaic result. MAIN OUTCOME MEASURES Rates of spontaneous abortion, birth weight, length of gestation, incidence of birth defects, and chromosomal status during gestation. RESULTS Implanted euploid embryos had a significantly lower risk of spontaneous abortion compared with mosaic embryos (8.9% [n = 8,672; 95% confidence interval {CI95} 8.3, 9.5] vs. 22.2% [n = 914; CI95 19.6, 25.0]). Embryos with mosaicism affecting whole chromosomes (not segmental) had the highest risk of spontaneous abortion (27.6% [n = 395; CI95 23.2, 32.3]). Infants born from euploid, mosaic, and whole chromosome mosaic embryos had average birth weights and lengths of gestation that were not statistically different (3,118 g and 267 days [n = 488; CI95 3,067, 3,169, and 266, 268], 3052 g and 265 days [n = 488; CI95 2,993, 3,112, and 264,267], 3,159 g and 268 days [n = 194; CI95 3,070, 3,249, and 266,270], respectively). Out of 488 infants from mosaic embryo transfers (ETs), one had overt gross abnormalities as defined by the Centers for Disease Control and Prevention. Most prenatal tests performed on pregnancies from mosaic ETs had normal results, and only three pregnancies produced prenatal test results reflecting the mosaicism detected at the embryonic stage (3 out of 250, 1.2%; CI95 0.25, 3.5). CONCLUSION Although embryos classified as mosaic experience higher rates of miscarriage than euploid embryos (with a particularly high frequency shortly after implantation), infants born of mosaic ETs are similar to infants of euploid ETs. Prenatal testing indicates that mosaicism resolves during most pregnancies, although this process is not perfectly efficient. In a small percentage of cases, the mosaicism persists through gestation. These findings can serve as risk-benefit considerations for mosaic ETs in the fertility clinic.
Collapse
Affiliation(s)
- Manuel Viotti
- Zouves Foundation for Reproductive Medicine, Foster City, California; Kindlabs, Kindbody, New York, New York.
| | - Ermanno Greco
- Villa Mafalda, Center For Reproductive Medicine, Rome, Italy
| | - James A Grifo
- New York University Langone Fertility Center, New York, New York
| | - Mitko Madjunkov
- CReATe Fertility Centre, Toronto, Canada; Department of Obstetrics and Gynecology, University of Toronto, Toronto, Canada
| | - Clifford Librach
- CReATe Fertility Centre, Toronto, Canada; Department of Obstetrics and Gynecology, University of Toronto, Toronto, Canada; Institute of Medical Sciences and Department of Physiology, University of Toronto, Toronto, Canada
| | | | | | - Pavel Yakovlev
- Centre for Reproductive Medicine, Co.Ltd. "Next Generation Clinic," Moscow, Russia
| | - Nikolay Kornilov
- Centre for Reproductive Medicine, Co.Ltd. "Next Generation Clinic," Moscow, Russia; Centre for Reproductive Medicine, Co.Ltd. "Next Generation Clinic," St. Petersburg, Russia
| | - Laura Corti
- IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Anil Biricik
- Eurofins Genoma Group, Molecular Genetics Laboratories, Rome, Italy
| | | | | | - Maw-Sheng Lee
- Lee Women's Hospital, Taichung, Taiwan; Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan
| | | | | | - Darren K Griffin
- School of Biosciences, University of Kent, Canterbury, United Kingdom
| | - Diane Y Tran
- Zouves Fertility Center, Foster City, California
| | - Purvi Kaur
- Zouves Fertility Center, Foster City, California
| | - Frank L Barnes
- Zouves Foundation for Reproductive Medicine, Foster City, California; Zouves Fertility Center, Foster City, California
| | - Christo G Zouves
- Zouves Foundation for Reproductive Medicine, Foster City, California; Zouves Fertility Center, Foster City, California
| | - Andrea R Victor
- School of Biosciences, University of Kent, Canterbury, United Kingdom; Zouves Fertility Center, Foster City, California; Reproductive Medicine Associates of Long Island, Melville, New York
| | - Andria G Besser
- New York University Langone Fertility Center, New York, New York
| | - Svetlana Madjunkova
- CReATe Fertility Centre, Toronto, Canada; Department of Obstetrics and Gynecology, University of Toronto, Toronto, Canada; Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Canada
| | | |
Collapse
|
33
|
McCoy RC, Summers MC, McCollin A, Ottolini CS, Ahuja K, Handyside AH. Meiotic and mitotic aneuploidies drive arrest of in vitro fertilized human preimplantation embryos. Genome Med 2023; 15:77. [PMID: 37779206 PMCID: PMC10544495 DOI: 10.1186/s13073-023-01231-1] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Accepted: 09/12/2023] [Indexed: 10/03/2023] Open
Abstract
BACKGROUND The high incidence of aneuploidy in early human development, arising either from errors in meiosis or postzygotic mitosis, is the primary cause of pregnancy loss, miscarriage, and stillbirth following natural conception as well as in vitro fertilization (IVF). Preimplantation genetic testing for aneuploidy (PGT-A) has confirmed the prevalence of meiotic and mitotic aneuploidies among blastocyst-stage IVF embryos that are candidates for transfer. However, only about half of normally fertilized embryos develop to the blastocyst stage in vitro, while the others arrest at cleavage to late morula or early blastocyst stages. METHODS To achieve a more complete view of the impacts of aneuploidy, we applied low-coverage sequencing-based PGT-A to a large series (n = 909) of arrested embryos and trophectoderm biopsies. We then correlated observed aneuploidies with abnormalities of the first two cleavage divisions using time-lapse imaging (n = 843). RESULTS The combined incidence of meiotic and mitotic aneuploidies was strongly associated with blastocyst morphological grading, with the proportion ranging from 20 to 90% for the highest to lowest grades, respectively. In contrast, the incidence of aneuploidy among arrested embryos was exceptionally high (94%), dominated by mitotic aneuploidies affecting multiple chromosomes. In turn, these mitotic aneuploidies were strongly associated with abnormal cleavage divisions, such that 51% of abnormally dividing embryos possessed mitotic aneuploidies compared to only 23% of normally dividing embryos. CONCLUSIONS We conclude that the combination of meiotic and mitotic aneuploidies drives arrest of human embryos in vitro, as development increasingly relies on embryonic gene expression at the blastocyst stage.
Collapse
Affiliation(s)
- Rajiv C McCoy
- Department of Biology, Johns Hopkins University, 3400 N. Charles Street, Baltimore, MD, 21212, USA.
| | - Michael C Summers
- London Women's Clinic, 113-115 Harley Street, Marylebone, London, W1G 6AP, UK
- School of Biosciences, University of Kent, Canterbury, CT2 7NJ, Kent, UK
- Present Address: London Women's Clinic, The Chesterfield, Nuffield Health Clinic, 3 Clifton Hill, Bristol, BS8 1BN, UK
| | - Abeo McCollin
- London Women's Clinic, 113-115 Harley Street, Marylebone, London, W1G 6AP, UK
- School of Biosciences, University of Kent, Canterbury, CT2 7NJ, Kent, UK
| | - Christian S Ottolini
- London Women's Clinic, 113-115 Harley Street, Marylebone, London, W1G 6AP, UK
- Department of Maternal and Fetal Medicine, University College London, 86-96 Chenies Mews, London, WC1E 6HX, UK
- Present Address: Juno Genetics Italia, Via Di Quarto Peperino 22, 00188, Rome, Italy
| | - Kamal Ahuja
- London Women's Clinic, 113-115 Harley Street, Marylebone, London, W1G 6AP, UK
| | - Alan H Handyside
- School of Biosciences, University of Kent, Canterbury, CT2 7NJ, Kent, UK
| |
Collapse
|
34
|
Ju LF, Xu HJ, Yang YG, Yang Y. Omics Views of Mechanisms for Cell Fate Determination in Early Mammalian Development. GENOMICS, PROTEOMICS & BIOINFORMATICS 2023; 21:950-961. [PMID: 37075831 PMCID: PMC10928378 DOI: 10.1016/j.gpb.2023.03.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/18/2023] [Revised: 03/18/2023] [Accepted: 03/23/2023] [Indexed: 04/21/2023]
Abstract
During mammalian preimplantation development, a totipotent zygote undergoes several cell cleavages and two rounds of cell fate determination, ultimately forming a mature blastocyst. Along with compaction, the establishment of apicobasal cell polarity breaks the symmetry of an embryo and guides subsequent cell fate choice. Although the lineage segregation of the inner cell mass (ICM) and trophectoderm (TE) is the first symbol of cell differentiation, several molecules have been shown to bias the early cell fate through their inter-cellular variations at much earlier stages, including the 2- and 4-cell stages. The underlying mechanisms of early cell fate determination have long been an important research topic. In this review, we summarize the molecular events that occur during early embryogenesis, as well as the current understanding of their regulatory roles in cell fate decisions. Moreover, as powerful tools for early embryogenesis research, single-cell omics techniques have been applied to both mouse and human preimplantation embryos and have contributed to the discovery of cell fate regulators. Here, we summarize their applications in the research of preimplantation embryos, and provide new insights and perspectives on cell fate regulation.
Collapse
Affiliation(s)
- Lin-Fang Ju
- Sino-Danish College, University of Chinese Academy of Sciences, Beijing 100049, China; University of Chinese Academy of Sciences, Beijing 100049, China; CAS Key Laboratory of Genomic and Precision Medicine, Collaborative Innovation Center of Genetics and Development, Beijing Institute of Genomics, Chinese Academy of Sciences and China National Center for Bioinformation, Beijing 100101, China
| | - Heng-Ji Xu
- University of Chinese Academy of Sciences, Beijing 100049, China; CAS Key Laboratory of Genomic and Precision Medicine, Collaborative Innovation Center of Genetics and Development, Beijing Institute of Genomics, Chinese Academy of Sciences and China National Center for Bioinformation, Beijing 100101, China
| | - Yun-Gui Yang
- Sino-Danish College, University of Chinese Academy of Sciences, Beijing 100049, China; University of Chinese Academy of Sciences, Beijing 100049, China; CAS Key Laboratory of Genomic and Precision Medicine, Collaborative Innovation Center of Genetics and Development, Beijing Institute of Genomics, Chinese Academy of Sciences and China National Center for Bioinformation, Beijing 100101, China; Institute of Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing 100101, China.
| | - Ying Yang
- Sino-Danish College, University of Chinese Academy of Sciences, Beijing 100049, China; University of Chinese Academy of Sciences, Beijing 100049, China; CAS Key Laboratory of Genomic and Precision Medicine, Collaborative Innovation Center of Genetics and Development, Beijing Institute of Genomics, Chinese Academy of Sciences and China National Center for Bioinformation, Beijing 100101, China; Institute of Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing 100101, China.
| |
Collapse
|
35
|
Orvieto R. Failure to detect DNA in blastocoel fluids after whole genome amplification-is it the next adds-on? Hum Reprod 2023; 38:1853-1855. [PMID: 37464894 DOI: 10.1093/humrep/dead140] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/20/2023] Open
Affiliation(s)
- Raoul Orvieto
- Infertility and IVF Institute, Department of Obstetrics and Gynecology, Chaim Sheba Medical Center (Tel Hashomer), Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| |
Collapse
|
36
|
Kirsch-Volders M, Fenech M. Towards prevention of aneuploidy-associated cellular senescence and aging: more questions than answers? MUTATION RESEARCH. REVIEWS IN MUTATION RESEARCH 2023; 792:108474. [PMID: 37866738 DOI: 10.1016/j.mrrev.2023.108474] [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: 02/15/2023] [Revised: 10/10/2023] [Accepted: 10/16/2023] [Indexed: 10/24/2023]
Abstract
The aim of this review is to discuss how aneuploidy contributes to the aging process, and to identify plausible strategies for its prevention. After an overview of mechanisms leading to aneuploidy and the major features of cellular senescence, we discuss the link between (i) aneuploidy and cellular senescence; (ii) aneuploidy and aging; and (iii) cellular senescence and aging. We also consider (i) interactions between aneuploidy, micronuclei, cellular senescence and aging, (ii) the potential of nutritional treatments to prevent aneuploidy-associated senescence and aging, and (iii) knowledge and technological gaps. Evidence for a causal link between aneuploidy, senescence and aging is emerging. In vitro, aneuploidy accompanies the entry into cellular senescence and can itself induce senescence. How aneuploidy contributes in vivo to cellular senescence is less clear. Several routes depending on aneuploidy and/or senescence converge towards chronic inflammation, the major driver of unhealthy aging. Aneuploidy can induce the pro-inflammatory Senescence Associated Secretory Phenotype (SASP), either directly or as a result of micronucleus (MN) induction leading to leakage of DNA into the cytoplasm and triggering of the cGAS-STING pathway of innate immune response. A major difficulty in understanding the impact of aneuploidy on senescence and aging in vivo, results from the heterogeneity of cellular senescence in different tissues at the cytological and molecular level. Due to this complexity, there is at the present time no biomarker or biomarker combination characteristic for all types of senescent cells. In conclusion, a deeper understanding of the critical role aneuploidy plays in cellular senescence and aging is essential to devise practical strategies to protect human populations from aneuploidy-associated pathologies. We discuss emerging evidence, based on in vitro and in vivo studies, that adequate amounts of specific micronutrients are essential for prevention of aneuploidy in humans and that precise nutritional intervention may be essential to help avoid the scourge of aneuploidy-driven diseases.
Collapse
Affiliation(s)
- Micheline Kirsch-Volders
- Laboratory for Cell Genetics, Department Biology, Faculty of Sciences and Bio-engineering Sciences, Vrije Universiteit Brussel, Pleinlaan 2, 1050 Brussels, Belgium.
| | - Michael Fenech
- Clinical and Health Sciences, University of South Australia, SA 5000, Australia; Genome Health Foundation, North Brighton, SA 5048, Australia.
| |
Collapse
|
37
|
Abstract
Embryo implantation in humans is interstitial, meaning the entire conceptus embeds in the endometrium before the placental trophoblast invades beyond the uterine mucosa into the underlying inner myometrium. Once implanted, embryo survival pivots on the transformation of the endometrium into an anti-inflammatory placental bed, termed decidua, under homeostatic control of uterine natural killer cells. Here, we examine the evolutionary context of embryo implantation and elaborate on uterine remodelling before and after conception in humans. We also discuss the interactions between the embryo and the decidualising endometrium that regulate interstitial implantation and determine embryo fitness. Together, this Review highlights the precarious but adaptable nature of the implantation process.
Collapse
Affiliation(s)
- Joanne Muter
- Division of Biomedical Sciences, Warwick Medical School, University of Warwick, Coventry, CV2 2DX, UK
- Tommy's National Centre for Miscarriage Research, University Hospitals Coventry & Warwickshire NHS Trust, Warwick Medical School, University of Warwick, Coventry, CV2 2DX, UK
| | - Vincent J. Lynch
- Department of Biological Sciences, University at Buffalo, Buffalo, NY 14260-4610, USA
| | - Rajiv C. McCoy
- Department of Biology, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Jan J. Brosens
- Division of Biomedical Sciences, Warwick Medical School, University of Warwick, Coventry, CV2 2DX, UK
- Tommy's National Centre for Miscarriage Research, University Hospitals Coventry & Warwickshire NHS Trust, Warwick Medical School, University of Warwick, Coventry, CV2 2DX, UK
| |
Collapse
|
38
|
Gleicher N, Mochizuki L, Barad DH, Patrizio P, Orvieto R. A review of the 2021/2022 PGDIS Position Statement on the transfer of mosaic embryos. J Assist Reprod Genet 2023; 40:817-826. [PMID: 36892704 PMCID: PMC10224891 DOI: 10.1007/s10815-023-02763-6] [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: 12/14/2022] [Accepted: 02/22/2023] [Indexed: 03/10/2023] Open
Abstract
The practice of preimplantation genetic testing for aneuploidy (PGT-A) in association with in vitro fertilization (IVF) since 2016 has been mostly directed by three highly controversial guidance documents issued by the Preimplantation Genetic Diagnosis International Society (PGDIS). Because these documents are so influential on worldwide IVF practice, the most recent one is here the subject of a detailed review, again revealing important misrepresentations and internal contradictions. Most importantly, however, this most recent guidance document still does not prevent the non-use and/or disposal of large numbers of embryos with substantial pregnancy and live-birth potential and, therefore, continues to propagate an IVF practice harmful to many infertile women.
Collapse
Affiliation(s)
- Norbert Gleicher
- The Center for Human Reproduction, New York, NY, USA.
- Foundation for Reproductive Medicine, New York, NY, USA.
- Stem Cell Biology and Molecular Embryology Laboratory, The Rockefeller University, New York, NY, USA.
- Department of Obstetrics and Gynecology, Medical University of Vienna, 1090, Vienna, Austria.
| | | | - David H Barad
- The Center for Human Reproduction, New York, NY, USA
- Foundation for Reproductive Medicine, New York, NY, USA
| | - Pasquale Patrizio
- The Center for Human Reproduction, New York, NY, USA
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of Miami, Miller School of Medicine, Miami, FL, USA
| | - Raoul Orvieto
- Chaim Sheba Medical Center, Infertility and IVF Unit, Department of Obstetrics and Gynecology, Tel Aviv University, Sackler Medical Faculty, Tel-Aviv, Israel
| |
Collapse
|
39
|
Dellatorre G, Fava VM, Mira MT, Silva de Castro CC. Experimental approaches to assess melanocytes mosaicism in segmental vitiligo. An Bras Dermatol 2023; 98:216-220. [PMID: 36529602 PMCID: PMC9984711 DOI: 10.1016/j.abd.2022.05.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Revised: 05/10/2022] [Accepted: 05/26/2022] [Indexed: 12/23/2022] Open
Abstract
Vitiligo is an autoimmune disease of the skin that results in localized or disseminated white macules. One common feature of several existing classification protocols is the distribution of the disease into two main subtypes, non-segmental vitiligo (NSV) and segmental vitiligo (SV). SV is characterized by depigmentation spreading within one or more skin segments while NSV is widespread. Several clinical-epidemiological observations suggest that SV has distinct autoimmune pathophysiology compared to NSV. Furthermore, the clinical distribution pattern of SV lesions closely resembles other melanocyte mosaicism diseases. These observations led us to hypothesize that SV is caused by a localized autoimmune reaction targeting epidermal mosaicism melanocytes. Here, we proposed examples of experimental approaches to assess mosaicism in SV patients.
Collapse
Affiliation(s)
| | - Vinicius M Fava
- Infectious Diseases and Immunity in Global Health Program, The Research Institute of the McGill University Health Centre, Montreal, QC, Canada
| | - Marcelo Távora Mira
- Santa Casa de Misericórdia Hospital, Curitiba, PR, Brazil; Graduate Program in Health Sciences, School of Medicine, Pontifícia Universidade Católica do Paraná, Curitiba, PR, Brazil; School of Life Sciences, Pontifícia Universidade Católica do Paraná, Curitiba, PR, Brazil
| | - Caio Cesar Silva de Castro
- Santa Casa de Misericórdia Hospital, Curitiba, PR, Brazil; School of Medicine, Pontifícia Universidade Católica do Paraná, Curitiba, PR, Brazil.
| |
Collapse
|
40
|
Ranisch R, Trettenbach K, Arnason G. Initial heritable genome editing: mapping a responsible pathway from basic research to the clinic. MEDICINE, HEALTH CARE, AND PHILOSOPHY 2023; 26:21-35. [PMID: 36414813 PMCID: PMC9984515 DOI: 10.1007/s11019-022-10115-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/27/2021] [Revised: 09/04/2022] [Accepted: 09/11/2022] [Indexed: 06/16/2023]
Abstract
Following the Second Summit on Human Gene Editing in Hong Kong in 2018, where the birth of two girls with germline genome editing was revealed, the need for a responsible pathway to the clinical application of human germline genome editing has been repeatedly emphasised. This paper aims to contribute to the ongoing discussion on research ethics issues in germline genome editing by exploring key issues related to the initial applications of CRISPR in reproductive medicine. Following an overview of the current discussion on bringing germline genome editing into clinical practice, we outline the specific challenges associated with such interventions and the features that distinguish them from conventional clinical testing of new medical treatments. We then review proposed ethical requirements for initial heritable genome editing, such as the absence of reasonable alternatives, the existence of sufficient and reliable preclinical data, appropriate informed consent, requirements related to safety, and long-term follow-up.
Collapse
Affiliation(s)
- Robert Ranisch
- Junior Professorship for Medical Ethics with a Focus on Digitization, Faculty of Health Sciences Brandenburg, University of Potsdam, Am Mühlberg 9, 14476, Potsdam, Golm, Germany.
- Research Unit "Ethics of Genome Editing", Institute of Ethics and History of Medicine, University of Tübingen, Gartenstraße 47, D-72074, Tübingen, Germany.
| | - Katharina Trettenbach
- Junior Professorship for Medical Ethics with a Focus on Digitization, Faculty of Health Sciences Brandenburg, University of Potsdam, Am Mühlberg 9, 14476, Potsdam, Golm, Germany
- Research Unit "Ethics of Genome Editing", Institute of Ethics and History of Medicine, University of Tübingen, Gartenstraße 47, D-72074, Tübingen, Germany
| | - Gardar Arnason
- Research Unit "Ethics of Genome Editing", Institute of Ethics and History of Medicine, University of Tübingen, Gartenstraße 47, D-72074, Tübingen, Germany
- University of Akureyri, Norðurslóð 2, 600, Akureyri, Iceland
| |
Collapse
|
41
|
Hernandez Mora JR, Buhigas C, Clark S, Del Gallego Bonilla R, Daskeviciute D, Monteagudo-Sánchez A, Poo-Llanillo ME, Medrano JV, Simón C, Meseguer M, Kelsey G, Monk D. Single-cell multi-omic analysis profiles defective genome activation and epigenetic reprogramming associated with human pre-implantation embryo arrest. Cell Rep 2023; 42:112100. [PMID: 36763500 DOI: 10.1016/j.celrep.2023.112100] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Revised: 10/14/2022] [Accepted: 01/26/2023] [Indexed: 02/11/2023] Open
Abstract
During pre-implantation stages of mammalian development, maternally stored material promotes both the erasure of the sperm and oocyte epigenetic profiles and is responsible for concomitant genome activation. Here, we have utilized single-cell methylome and transcriptome sequencing (scM&T-seq) to quantify both mRNA expression and DNA methylation in oocytes and a developmental series of human embryos at single-cell resolution. We fully characterize embryonic genome activation and maternal transcript degradation and map key epigenetic reprogramming events in developmentally high-quality embryos. By comparing these signatures with early embryos that have undergone spontaneous cleavage-stage arrest, as determined by time-lapse imaging, we identify embryos that fail to appropriately activate their genomes or undergo epigenetic reprogramming. Our results indicate that a failure to successfully accomplish these essential milestones impedes the developmental potential of pre-implantation embryos and is likely to have important implications, similar to aneuploidy, for the success of assisted reproductive cycles.
Collapse
Affiliation(s)
| | - Claudia Buhigas
- Biomedical Research Centre, School of Biological Sciences, University of East Anglia, Norwich Research Park, Norwich NR4 7JT, UK
| | - Stephen Clark
- The Babraham Institute, Babraham, Cambridge CB22 3AT, UK
| | | | - Dagne Daskeviciute
- Biomedical Research Centre, School of Biological Sciences, University of East Anglia, Norwich Research Park, Norwich NR4 7JT, UK
| | | | | | - Jose Vicente Medrano
- IVI-RMA Global and Health Research Institute la Fe, 46026 Valencia, Spain; Department of Obstetrics and Gynecology, Valencia University and INCLIVA, 46010 Valencia, Spain
| | - Carlos Simón
- Department of Obstetrics and Gynecology, Valencia University and INCLIVA, 46010 Valencia, Spain; Department of Obstetrics and Gynecology, BIDMC, Harvard University, Boston, MA 02215, USA
| | - Marcos Meseguer
- IVI-RMA Global and Health Research Institute la Fe, 46026 Valencia, Spain
| | - Gavin Kelsey
- The Babraham Institute, Babraham, Cambridge CB22 3AT, UK; Centre for Trophoblast Research, University of Cambridge, Cambridge CB2 3EL, UK; Wellcome-MRC Institute of Metabolic Science-Metabolic Research Laboratories, Cambridge CB2 0QQ, UK
| | - David Monk
- Bellvitge Institute for Biomedical Research, 08908 Barcelona, Spain; Biomedical Research Centre, School of Biological Sciences, University of East Anglia, Norwich Research Park, Norwich NR4 7JT, UK.
| |
Collapse
|
42
|
The Human Early Maternal–Embryonic Interactome. REPRODUCTIVE MEDICINE 2023. [DOI: 10.3390/reprodmed4010006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/18/2023] Open
Abstract
Background: Single cell transcriptomics offers an avenue for predicting, with improved accuracy, the gene networks that are involved in the establishment of the first direct cell–cell interactions between the blastocyst and the maternal luminal epithelium. We hypothesised that in silico modelling of the maternal–embryonic interface may provide a causal model of these interactions, leading to the identification of genes associated with a successful initiation of implantation. Methods: Bulk and single cell RNA-sequencing of endometrial epithelium and scRNAseq of day 6 and 7 trophectoderm (TE) were used to model the initial encounter between the blastocyst and the maternal uterine lining epithelium in silico. In silico modelling of the maternal–embryonic interface was performed using hypernetwork (HN) analysis of genes mediating endometrial–TE interactions and the wider endometrial epithelial transcriptome. A hypernetwork analysis identifies genes that co-ordinate the expression of many other genes to derive a higher order interaction likely to be causally linked to the function. Potential interactions of TE with non-ciliated luminal cells, ciliated cells, and glandular cells were examined. Results: Prominent epithelial activities include secretion, endocytosis, ion transport, adhesion, and immune modulation. Three highly correlated clusters of 25, 22 and 26 TE-interacting epithelial surface genes were identified, each with distinct properties. Genes in both ciliated and non-ciliated luminal epithelial cells and glandular cells exhibit significant functional associations. Ciliated cells are predicted to bind to TE via galectin–glycan interaction. Day 6 and day 7 embryonic–epithelial interactomes are largely similar. The removal of aneuploid TE-derived mRNA invoked only subtle differences. No direct interaction with the maternal gland epithelial cell surface is predicted. These functional differences validate the in silico segregation of phenotypes. Single cell analysis of the epithelium revealed significant change with the cycle phase, but differences in the cell phenotype between individual donors were also present. Conclusions: A hypernetwork analysis can identify epithelial gene clusters that show correlated change during the menstrual cycle and can be interfaced with TE genes to predict pathways and processes occurring during the initiation of embryo–epithelial interaction in the mid-secretory phase. The data are on a scale that is realistic for functional dissection using current ex vivo human implantation models. A focus on luminal epithelial cells may allow a resolution to the current bottleneck of endometrial receptivity testing based on tissue lysates, which is confounded by noise from multiple diverse cell populations.
Collapse
|
43
|
Deng C, Ya A, Compton DA, Godek KM. A pluripotent developmental state confers a low fidelity of chromosome segregation. Stem Cell Reports 2023; 18:475-488. [PMID: 36638786 PMCID: PMC9968987 DOI: 10.1016/j.stemcr.2022.12.008] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 12/09/2022] [Accepted: 12/12/2022] [Indexed: 01/13/2023] Open
Abstract
During in vitro propagation, human pluripotent stem cells (hPSCs) frequently become aneuploid with incorrect chromosome numbers due to mitotic chromosome segregation errors. Yet, it is not understood why hPSCs exhibit a low mitotic fidelity. Here, we investigate the mechanisms responsible for mitotic errors in hPSCs and show that the primary cause is lagging chromosomes in anaphase with improper merotelic microtubule attachments. Accordingly, short-term treatment (<24 h) with small molecules that prolong mitotic duration or destabilize chromosome microtubule attachments reduces merotelic errors and lagging chromosome rates, although hPSCs adapt and lagging chromosome rates rebound upon long-term (>24 h) microtubule destabilization. Strikingly, we also demonstrate that mitotic error rates correlate with developmental potential decreasing or increasing upon loss or gain of pluripotency, respectively. Thus, a low mitotic fidelity is an inherent and conserved phenotype of hPSCs. Moreover, chromosome segregation fidelity depends on developmental state in normal human cells.
Collapse
Affiliation(s)
- Chenhui Deng
- Department of Biochemistry and Cell Biology, Geisel School of Medicine at Dartmouth, Hanover, NH, USA; Dartmouth Cancer Center, Geisel School of Medicine at Dartmouth, Lebanon, NH, USA
| | - Amanda Ya
- Department of Biochemistry and Cell Biology, Geisel School of Medicine at Dartmouth, Hanover, NH, USA; Dartmouth Cancer Center, Geisel School of Medicine at Dartmouth, Lebanon, NH, USA
| | - Duane A Compton
- Department of Biochemistry and Cell Biology, Geisel School of Medicine at Dartmouth, Hanover, NH, USA; Dartmouth Cancer Center, Geisel School of Medicine at Dartmouth, Lebanon, NH, USA
| | - Kristina M Godek
- Department of Biochemistry and Cell Biology, Geisel School of Medicine at Dartmouth, Hanover, NH, USA; Dartmouth Cancer Center, Geisel School of Medicine at Dartmouth, Lebanon, NH, USA.
| |
Collapse
|
44
|
Prasad D, Illek K, Fischer F, Holstein K, Classen AK. Bilateral JNK activation is a hallmark of interface surveillance and promotes elimination of aberrant cells. eLife 2023; 12:e80809. [PMID: 36744859 PMCID: PMC9917460 DOI: 10.7554/elife.80809] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2022] [Accepted: 02/03/2023] [Indexed: 02/07/2023] Open
Abstract
Tissue-intrinsic defense mechanisms eliminate aberrant cells from epithelia and thereby maintain the health of developing tissues or adult organisms. 'Interface surveillance' comprises one such distinct mechanism that specifically guards against aberrant cells which undergo inappropriate cell fate and differentiation programs. The cellular mechanisms which facilitate detection and elimination of these aberrant cells are currently unknown. We find that in Drosophila imaginal discs, clones of cells with inappropriate activation of cell fate programs induce bilateral JNK activation at clonal interfaces, where wild type and aberrant cells make contact. JNK activation is required to drive apoptotic elimination of interface cells. Importantly, JNK activity and apoptosis are highest in interface cells within small aberrant clones, which likely supports the successful elimination of aberrant cells when they arise. Our findings are consistent with a model where clone size affects the topology of interface contacts and thereby the strength of JNK activation in wild type and aberrant interface cells. Bilateral JNK activation is unique to 'interface surveillance' and is not observed in other tissue-intrinsic defense mechanisms, such as classical 'cell-cell competition'. Thus, bilateral JNK interface signaling provides an independent tissue-level mechanism to eliminate cells with inappropriate developmental fate but normal cellular fitness. Finally, oncogenic Ras-expressing clones activate 'interface surveillance' but evade elimination by bilateral JNK activation. Combined, our work establishes bilateral JNK interface signaling and interface apoptosis as a new hallmark of interface surveillance and highlights how oncogenic mutations evade tumor suppressor function encoded by this tissue-intrinsic surveillance system.
Collapse
Affiliation(s)
- Deepti Prasad
- Hilde-Mangold-Haus, University of FreiburgFreiburgGermany
- Spemann Graduate School of Biology and Medicine (SGBM), University of FreiburgFreiburgGermany
- Faculty of Biology, University of FreiburgFreiburgGermany
| | | | - Friedericke Fischer
- Hilde-Mangold-Haus, University of FreiburgFreiburgGermany
- Faculty of Biology, University of FreiburgFreiburgGermany
- International Max Planck Research School for Immunobiology, Epigenetics, and MetabolismFreiburgGermany
| | | | - Anne-Kathrin Classen
- Hilde-Mangold-Haus, University of FreiburgFreiburgGermany
- Faculty of Biology, University of FreiburgFreiburgGermany
- CIBSS Centre for Integrative Biological Signalling Studies, University of FreiburgFreiburgGermany
- BIOSS Centre for Biological Signalling Studies, University of FreiburgFreiburgGermany
| |
Collapse
|
45
|
Greco E, Yakovlev P, Kornilov N, Vyatkina S, Bogdanova D, Ermakova M, Tarasova Y, Tikhonov A, Pendina A, Biricik A, Sessa MT, Listorti I, Ronsini C, Greco PF, Victor A, Barnes F, Zouves C, Spinella F, Viotti M. Two clinical case reports of embryonic mosaicism identified with PGT-A persisting during pregnancy as true fetal mosaicism. Hum Reprod 2023; 38:315-323. [PMID: 36610460 DOI: 10.1093/humrep/deac263] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2022] [Revised: 10/31/2022] [Indexed: 01/09/2023] Open
Abstract
The health risks associated with transferring embryos classified as mosaic by preimplantation genetic testing for aneuploidies (PGT-A) are currently unknown. Such embryos produce PGT-A results indicating the presence of both euploid and aneuploid cells and have historically been deselected from transfer and grouped with uniformly aneuploid embryos as 'abnormal'. In recent years, numerous groups have reported the intentional transfer of mosaic embryos in the absence of uniformly euploid embryos, largely observing births of seemingly healthy babies. However, it remains to be understood whether the embryonic mosaicism invariably becomes resolved during the ensuing pregnancy, or whether the placenta and/or fetal tissues retain aneuploid cells, and if so to what potential clinical effect. Here, we report two cases of mosaicism persisting from the embryonic stage to the established pregnancy. Case 1 involved an embryonic low-level segmental mosaic loss in Chromosome (Chr) 1, which was confirmed in amniocentesis as well as in brain tissue of the products of conception. This pregnancy was terminated due to the chromosomal pathologies associated with 1p36 deletion syndrome, such as severe intellectual disability. Case 2 involved a low-level mosaic Chr 21 trisomy, which was confirmed with chorionic villus sampling and amniocentesis. The ensuing pregnancy was terminated after ultrasound identification of severe abnormalities in the placenta and fetus. Together, these two cases should be taken into account for risk-benefit assessments of prospective mosaic embryo transfers.
Collapse
Affiliation(s)
- Ermanno Greco
- Villa Mafalda, Centre For Reproductive Medicine, Rome, Italy.,Department of Obstetrics and Gynecology, UniCamillus International University, Rome, Italy
| | - Pavel Yakovlev
- Next Generation Clinic, Centre For Reproductive Medicine, Moscow, Russia
| | - Nikolay Kornilov
- Next Generation Clinic, Centre For Reproductive Medicine, Moscow, Russia.,Next Generation Clinic, Centre For Reproductive Medicine, St. Petersburg, Russia
| | - Svetlana Vyatkina
- Next Generation Clinic, Centre For Reproductive Medicine, St. Petersburg, Russia
| | - Daria Bogdanova
- Next Generation Clinic, Centre For Reproductive Medicine, Moscow, Russia
| | - Marina Ermakova
- Medical Genetic Center of the Group of companies "Mother and Child", Moscow, Russia
| | - Yulia Tarasova
- Medical Genetic Center of the Group of companies "Mother and Child", Moscow, Russia
| | - Andrei Tikhonov
- D.O. Ott Research Institute of Obstetrics, Gynecology and Reproduction, Saint Petersburg, Russia
| | - Anna Pendina
- D.O. Ott Research Institute of Obstetrics, Gynecology and Reproduction, Saint Petersburg, Russia
| | - Anil Biricik
- Eurofins Genoma Group, Molecular Genetics Laboratories, Rome, Italy
| | | | - Ilaria Listorti
- Villa Mafalda, Centre For Reproductive Medicine, Rome, Italy
| | - Carlo Ronsini
- Dipartimento della Donna, del Bambino e di Chirurgia Generale e Specialistica, University of Campania Luigi Vanvitelli, Naples, Italy
| | | | | | | | | | | | - Manuel Viotti
- Zouves Fertility Center, Foster City, CA, USA.,Zouves Foundation for Reproductive Medicine, Foster City, CA, USA
| |
Collapse
|
46
|
Gravholt CH, Viuff M, Just J, Sandahl K, Brun S, van der Velden J, Andersen NH, Skakkebaek A. The Changing Face of Turner Syndrome. Endocr Rev 2023; 44:33-69. [PMID: 35695701 DOI: 10.1210/endrev/bnac016] [Citation(s) in RCA: 65] [Impact Index Per Article: 32.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Indexed: 01/20/2023]
Abstract
Turner syndrome (TS) is a condition in females missing the second sex chromosome (45,X) or parts thereof. It is considered a rare genetic condition and is associated with a wide range of clinical stigmata, such as short stature, ovarian dysgenesis, delayed puberty and infertility, congenital malformations, endocrine disorders, including a range of autoimmune conditions and type 2 diabetes, and neurocognitive deficits. Morbidity and mortality are clearly increased compared with the general population and the average age at diagnosis is quite delayed. During recent years it has become clear that a multidisciplinary approach is necessary toward the patient with TS. A number of clinical advances has been implemented, and these are reviewed. Our understanding of the genomic architecture of TS is advancing rapidly, and these latest developments are reviewed and discussed. Several candidate genes, genomic pathways and mechanisms, including an altered transcriptome and epigenome, are also presented.
Collapse
Affiliation(s)
- Claus H Gravholt
- Department of Endocrinology and Internal Medicine, Aarhus University Hospital, Aarhus 8200 N, Denmark.,Department of Molecular Medicine, Aarhus University Hospital, Aarhus 8200 N, Denmark
| | - Mette Viuff
- Department of Endocrinology and Internal Medicine, Aarhus University Hospital, Aarhus 8200 N, Denmark.,Department of Molecular Medicine, Aarhus University Hospital, Aarhus 8200 N, Denmark
| | - Jesper Just
- Department of Molecular Medicine, Aarhus University Hospital, Aarhus 8200 N, Denmark
| | - Kristian Sandahl
- Department of Endocrinology and Internal Medicine, Aarhus University Hospital, Aarhus 8200 N, Denmark
| | - Sara Brun
- Department of Endocrinology and Internal Medicine, Aarhus University Hospital, Aarhus 8200 N, Denmark
| | - Janielle van der Velden
- Department of Pediatrics, Radboud University Medical Centre, Amalia Children's Hospital, 6525 Nijmegen, the Netherlands
| | - Niels H Andersen
- Department of Cardiology, Aalborg University Hospital, Aalborg 9000, Denmark
| | - Anne Skakkebaek
- Department of Molecular Medicine, Aarhus University Hospital, Aarhus 8200 N, Denmark.,Department of Clinical Genetics, Aarhus University Hospital, Aarhus 8200 N, Denmark
| |
Collapse
|
47
|
Griffin DK, Brezina PR, Tobler K, Zhao Y, Silvestri G, Mccoy RC, Anchan R, Benner A, Cutting GR, Kearns WG. The human embryonic genome is karyotypically complex, with chromosomally abnormal cells preferentially located away from the developing fetus. Hum Reprod 2023; 38:180-188. [PMID: 36350568 PMCID: PMC10089293 DOI: 10.1093/humrep/deac238] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Revised: 09/26/2022] [Indexed: 11/11/2022] Open
Abstract
STUDY QUESTION Are chromosome abnormalities detected at Day 3 post-fertilization predominantly retained in structures of the blastocyst other than the inner cell mass (ICM), where chromosomally normal cells are preferentially retained? SUMMARY ANSWER In human embryos, aneuploid cells are sequestered away from the ICM, partly to the trophectoderm (TE) but more significantly to the blastocoel fluid within the blastocoel cavity (Bc) and to peripheral cells (PCs) surrounding the blastocyst during Day 3 to Day 5 progression. WHAT IS KNOWN ALREADY A commonly held dogma in all diploid eukaryotes is that two gametes, each with 'n' chromosomes (23 in humans), fuse to form a '2n' zygote (46 in humans); a state that remains in perpetuity for all somatic cell divisions. Human embryos, however, display high levels of chromosomal aneuploidy in early stages that reportedly declines from Day 3 (cleavage stage) to Day 5 (blastocyst) post-fertilization. While this observation may be partly because of aneuploid embryonic arrest before blastulation, it could also be due to embryo 'normalization' to a euploid state during blastulation. If and how this normalization occurs requires further investigation. STUDY DESIGN, SIZE, DURATION A total of 964 cleavage-stage (Day 3) embryos underwent single-cell biopsy and diagnosis for chromosome constitution. All were maintained in culture, assessing blastulation rate, both for those assessed euploid and aneuploid. Pregnancy rate was assessed for those determined euploid, blastulated and subsequently transferred. For those determined aneuploid and blastulated (174 embryos), ICM (all 174 embryos), TE (all 174), Bc (47 embryos) and PC (38 embryos) were analyzed for chromosome constitution. Specifically, concordance with the original Day 3 diagnosis and determination if any 'normalized' to euploid karyotypes within all four structures was assessed. PARTICIPANTS/MATERIALS, SETTING, METHODS All patients (144 couples) were undergoing routine preimplantation genetic testing for aneuploidy in three IVF clinical settings. Cleavage-stage biopsy preceded chromosome analysis by next-generation sequencing. All patients provided informed consent. Additional molecular testing was carried out on blastocyst embryos and was analyzed for up to four embryonic structures (ICM, TE, Bc and PC). MAIN RESULTS AND THE ROLE OF CHANCE Of 463/964 embryos (48%) diagnosed as euploid at Day 3, 70% blastulated (leading to a 59% pregnancy rate) and 30% degenerated. Conversely, of the 501 (52%) diagnosed as aneuploid, 65% degenerated and 35% (174) blastulated, a highly significant difference (P < 0.0001). Of the 174 that blastulated, the ratio of '(semi)concordant-aneuploid' versus 'normalized-euploid' versus 'other-aneuploid' embryos was, respectively, 39%/57%/3% in the ICM; 49%/48%/3% in the TE; 78%/21%/0% in the PC; and 83%/10%/5% in the Bc. The TE karyotype therefore has a positive predictive value of 86.7% in determining that of the ICM, albeit with marginally higher aneuploid rates of abnormalities (P = .071). Levels of abnormality in Bc/PC were significantly higher (P < 0.0001) versus the ploidy of the ICM and TE and nearly all chromosome abnormalities were (at least partially) concordant with Day 3 diagnoses. LIMITATIONS, REASONS FOR CAUTION The results only pertain to human IVF embryos so extrapolation to the in vivo situation and to other species is not certain. We acknowledge (rather than lineage-specific survival, as we suggest here) the possibility of other mechanisms, such as lineage-specific movement of cells, during blastulation. Ethical considerations, however, make investigating this mechanism difficult on human embryos. WIDER IMPLICATIONS OF THE FINDINGS Mosaic human cleavage-stage embryos can differentiate into a euploid ICM where euploid cell populations predominate. Sequestering of aneuploid cells/nuclei to structures no longer involved in fetal development has important implications for preimplantation and prenatal genetic testing. These results also challenge previous fundamental understandings of mitotic fidelity in early human development and indicate a complex and fluid nature of the human embryonic genome. STUDY FUNDING/COMPETING INTEREST(S) This research was funded by Organon Pharmaceuticals and Merck Serono by grants to W.G.K. W.G.K. is also an employee of AdvaGenix, who could, potentially, indirectly benefit financially from publication of this manuscript. R.C.M. is supported by the National Institute of General Medical Sciences of the National Institutes of Health under award number R35GM133747. The content is solely the responsibility of the authors and does not necessarily represent the official views of the National Institutes of Health. D.K.G. provides paid consultancy services for Care Fertility. TRIAL REGISTRATION NUMBER : N/A.
Collapse
Affiliation(s)
- D K Griffin
- School of Biosciences, University of Kent, Canterbury, UK
| | - P R Brezina
- Jones Division of Reproductive Endocrinology, Department of Gynecology and Obstetrics, Johns Hopkins University School of Medicine, Baltimore, MD, USA.,Fertility Associates of Memphis, Memphis, TN, USA
| | - K Tobler
- Jones Division of Reproductive Endocrinology, Department of Gynecology and Obstetrics, Johns Hopkins University School of Medicine, Baltimore, MD, USA.,Idaho Center for Reproductive Medicine, Boise, ID, USA
| | - Yulian Zhao
- Jones Division of Reproductive Endocrinology, Department of Gynecology and Obstetrics, Johns Hopkins University School of Medicine, Baltimore, MD, USA.,Mayo Clinic, Rochester, MN, USA
| | - G Silvestri
- School of Biosciences, University of Kent, Canterbury, UK
| | - R C Mccoy
- Department of Biology, Johns Hopkins University, Baltimore, MD, USA
| | - R Anchan
- Department of Obstetrics and Gynecology, Harvard Medical School, Boston, MA, USA
| | | | - G R Cutting
- Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - W G Kearns
- Jones Division of Reproductive Endocrinology, Department of Gynecology and Obstetrics, Johns Hopkins University School of Medicine, Baltimore, MD, USA.,AdvaGenix, Rockville, MD, USA
| |
Collapse
|
48
|
Martin A, Mercader A, Dominguez F, Quiñonero A, Perez M, Gonzalez-Martin R, Delgado A, Mifsud A, Pellicer A, De Los Santos MJ. Mosaic results after preimplantation genetic testing for aneuploidy may be accompanied by changes in global gene expression. Front Mol Biosci 2023; 10:1180689. [PMID: 37122560 PMCID: PMC10140421 DOI: 10.3389/fmolb.2023.1180689] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Accepted: 04/04/2023] [Indexed: 05/02/2023] Open
Abstract
Aneuploidy in preimplantation embryos is a major cause of human reproductive failure. Unlike uniformly aneuploid embryos, embryos diagnosed as diploid-aneuploid mosaics after preimplantation genetic testing for aneuploidy (PGT-A) can develop into healthy infants. However, the reason why these embryos achieve full reproductive competence needs further research. Current RNA sequencing techniques allow for the investigation of the human preimplantation transcriptome, providing new insights into the molecular mechanisms of embryo development. In this prospective study, using euploid embryo gene expression as a control, we compared the transcriptome profiles of inner cell mass and trophectoderm samples from blastocysts with different levels of chromosomal mosaicism. A total of 25 samples were analyzed from 14 blastocysts with previous PGT-A diagnosis, including five low-level mosaic embryos and four high-level mosaic embryos. Global gene expression profiles visualized in cluster heatmaps were correlated with the original PGT-A diagnosis. In addition, gene expression distance based on the number of differentially expressed genes increased with the mosaic level, compared to euploid controls. Pathways involving apoptosis, mitosis, protein degradation, metabolism, and mitochondrial energy production were among the most deregulated within mosaic embryos. Retrospective analysis of the duration of blastomere cell cycles in mosaic embryos revealed several mitotic delays compared to euploid controls, providing additional evidence of the mosaic status. Overall, these findings suggest that embryos with mosaic results are not simply a misdiagnosis by-product, but may also have a genuine molecular identity that is compatible with their reproductive potential.
Collapse
Affiliation(s)
- A. Martin
- IVI-RMA Foundation, Health Research Institute La Fe, Valencia, Spain
| | - A. Mercader
- IVI-RMA Foundation, Health Research Institute La Fe, Valencia, Spain
- IVI-RMA Valencia, Valencia, Spain
| | - F. Dominguez
- IVI-RMA Foundation, Health Research Institute La Fe, Valencia, Spain
| | - A. Quiñonero
- IVI-RMA Foundation, Health Research Institute La Fe, Valencia, Spain
| | - M. Perez
- IVI-RMA Foundation, Health Research Institute La Fe, Valencia, Spain
| | | | | | | | - A. Pellicer
- IVI-RMA Foundation, Health Research Institute La Fe, Valencia, Spain
- IVI-RMA Rome, Rome, Italy
| | - M. J. De Los Santos
- IVI-RMA Foundation, Health Research Institute La Fe, Valencia, Spain
- IVI-RMA Valencia, Valencia, Spain
- *Correspondence: M. J. De Los Santos,
| |
Collapse
|
49
|
Kakourou G, Mamas T, Vrettou C, Traeger-Synodinos J. An Update on Non-invasive Approaches for Genetic Testing of the Preimplantation Embryo. Curr Genomics 2022; 23:337-352. [PMID: 36778192 PMCID: PMC9878856 DOI: 10.2174/1389202923666220927111158] [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: 04/20/2022] [Revised: 08/29/2022] [Accepted: 09/06/2022] [Indexed: 11/22/2022] Open
Abstract
Preimplantation Genetic Testing (PGT) aims to reduce the chance of an affected pregnancy or improve success in an assisted reproduction cycle. Since the first established pregnancies in 1990, methodological approaches have greatly evolved, combined with significant advances in the embryological laboratory. The application of preimplantation testing has expanded, while the accuracy and reliability of monogenic and chromosomal analysis have improved. The procedure traditionally employs an invasive approach to assess the nucleic acid content of embryos. All biopsy procedures require high technical skill, and costly equipment, and may impact both the accuracy of genetic testing and embryo viability. To overcome these limitations, many researchers have focused on the analysis of cell-free DNA (cfDNA) at the preimplantation stage, sampled either from the blastocoel or embryo culture media, to determine the genetic status of the embryo non-invasively. Studies have assessed the origin of cfDNA and its application in non-invasive testing for monogenic disease and chromosomal aneuploidies. Herein, we discuss the state-of-the-art for modern non-invasive embryonic genetic material assessment in the context of PGT. The results are difficult to integrate due to numerous methodological differences between the studies, while further work is required to assess the suitability of cfDNA analysis for clinical application.
Collapse
Affiliation(s)
- Georgia Kakourou
- Laboratory of Medical Genetics, National and Kapodistrian University of Athens, St. Sophia's Children's Hospital, 11527, Athens, Greece,Address correspondence to this author at the Laboratory of Medical Genetics, National and Kapodistrian University of Athens, St. Sophia's Children's Hospital, 11527, Athens, Greece; Tel/Fax: +302107467467; E-mail:
| | - Thalia Mamas
- Laboratory of Medical Genetics, National and Kapodistrian University of Athens, St. Sophia's Children's Hospital, 11527, Athens, Greece
| | - Christina Vrettou
- Laboratory of Medical Genetics, National and Kapodistrian University of Athens, St. Sophia's Children's Hospital, 11527, Athens, Greece
| | - Joanne Traeger-Synodinos
- Laboratory of Medical Genetics, National and Kapodistrian University of Athens, St. Sophia's Children's Hospital, 11527, Athens, Greece
| |
Collapse
|
50
|
Klaasen SJ, Kops GJPL. Chromosome Inequality: Causes and Consequences of Non-Random Segregation Errors in Mitosis and Meiosis. Cells 2022; 11:3564. [PMID: 36428993 PMCID: PMC9688425 DOI: 10.3390/cells11223564] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Revised: 11/01/2022] [Accepted: 11/08/2022] [Indexed: 11/16/2022] Open
Abstract
Aneuploidy is a hallmark of cancer and a major cause of miscarriages in humans. It is caused by chromosome segregation errors during cell divisions. Evidence is mounting that the probability of specific chromosomes undergoing a segregation error is non-random. In other words, some chromosomes have a higher chance of contributing to aneuploid karyotypes than others. This could have important implications for the origins of recurrent aneuploidy patterns in cancer and developing embryos. Here, we review recent progress in understanding the prevalence and causes of non-random chromosome segregation errors in mammalian mitosis and meiosis. We evaluate its potential impact on cancer and human reproduction and discuss possible research avenues.
Collapse
Affiliation(s)
- Sjoerd J. Klaasen
- Hubrecht Institute—KNAW (Royal Netherlands Academy of Arts and Sciences) and University Medical Centre Utrecht, 3584 CT Utrecht, The Netherlands
- Oncode Institute, 3521 AL Utrecht, The Netherlands
| | - Geert J. P. L. Kops
- Hubrecht Institute—KNAW (Royal Netherlands Academy of Arts and Sciences) and University Medical Centre Utrecht, 3584 CT Utrecht, The Netherlands
- Oncode Institute, 3521 AL Utrecht, The Netherlands
| |
Collapse
|