1
|
del Arco de la Paz A, Giménez-Rodríguez C, Selntigia A, Meseguer M, Galliano D. Advancements and Challenges in Preimplantation Genetic Testing for Aneuploidies: In the Pathway to Non-Invasive Techniques. Genes (Basel) 2024; 15:1613. [PMID: 39766880 PMCID: PMC11675356 DOI: 10.3390/genes15121613] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2024] [Revised: 12/08/2024] [Accepted: 12/12/2024] [Indexed: 01/11/2025] Open
Abstract
The evolution of preimplantation genetic testing for aneuploidy (PGT-A) techniques has been crucial in assisted reproductive technologies (ARTs), improving embryo selection and increasing success rates in in vitro fertilization (IVF) treatments. Techniques ranging from fluorescence in situ hybridization (FISH) to next-generation sequencing (NGS) have relied on cellular material extraction through biopsies of blastomeres at the cleavage stage on day three or from trophectoderm (TE) cells of the blastocyst. However, this has raised concerns about its potential impact on embryo development. As a result, there has been growing interest in developing non-invasive techniques for detecting aneuploidies, such as the analysis of blastocoel fluid (BF), spent culture medium (SCM), and artificial intelligence (AI) models. Non-invasive methods represent a promising advancement in PGT-A, offering the ability to detect aneuploidies without compromising embryo viability. This article reviews the evolution and principles of PGT-A, analyzing both traditional techniques and emerging non-invasive approaches, while highlighting the advantages and challenges associated with these methodologies. Furthermore, it explores the transformative potential of these innovations, which could optimize genetic screening and significantly improve clinical outcomes in the field of assisted reproduction.
Collapse
Affiliation(s)
- Ana del Arco de la Paz
- IVIRMA Global Research Alliance, IVI Foundation, Instituto de Investigación Sanitaria La Fe (IIS La Fe), 46026 Valencia, Spain
- IVIRMA Global Research Alliance, IVIRMA Valencia, 46015 Valencia, Spain
| | - Carla Giménez-Rodríguez
- IVIRMA Global Research Alliance, IVI Foundation, Instituto de Investigación Sanitaria La Fe (IIS La Fe), 46026 Valencia, Spain
- IVIRMA Global Research Alliance, IVIRMA Valencia, 46015 Valencia, Spain
| | | | - Marcos Meseguer
- IVIRMA Global Research Alliance, IVI Foundation, Instituto de Investigación Sanitaria La Fe (IIS La Fe), 46026 Valencia, Spain
- IVIRMA Global Research Alliance, IVIRMA Valencia, 46015 Valencia, Spain
| | | |
Collapse
|
2
|
Muñoz E, Bronet F, Lledo B, Palacios-Verdú G, Martinez-Rocca L, Altmäe S, Pla J. To transfer or not to transfer: the dilemma of mosaic embryos - a narrative review. Reprod Biomed Online 2024; 48:103664. [PMID: 38408811 DOI: 10.1016/j.rbmo.2023.103664] [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: 06/06/2023] [Revised: 10/19/2023] [Accepted: 10/30/2023] [Indexed: 02/28/2024]
Abstract
A frequent finding after preimplantation genetic diagnostic testing for aneuploidies using next-generation sequencing is an embryo that is putatively mosaic. The prevalence of this outcome remains unclear and varies with technical and external factors. Mosaic embryos can be classified by the percentage of cells affected, type of chromosome involvement (whole or segmental), number of affected chromosomes or affected cell type (inner mass cell, trophectoderm or both). The origin of mosaicism seems to be intrinsic as a post-zygotic mitotic error, but some external factors can play a role. As experience has increased with the transfer of mosaic embryos, clinical practice has gradually become more flexible in recent years. Nevertheless, clinical results show lower implantation, pregnancy and clinical pregnancy rates and higher miscarriage rates with mosaic embryo transfer when compared with the transfer of euploid embryos. Prenatal diagnosis is highly recommended after the transfer of mosaic embryos. This narrative review is intended to serve as reference material for practitioners in reproductive medicine who must manage a mosaic embryo result after preimplantation genetic testing for aneuploidies.
Collapse
Affiliation(s)
- Elkin Muñoz
- Reproductive Medicine, IVIRMA Vigo, Vigo, Spain; Department of Obstetrics and Gynecology, University of Cauca, Popayan, Colombia
| | | | | | - Gabriela Palacios-Verdú
- Unit of Genomic Medicine, Department of Obstetrics, Gynecology and Reproductive Medicine, Institut Universitari Quirón Dexeus, Barcelona, Spain
| | | | - Signe Altmäe
- Department of Biochemistry and Molecular Biology, Faculty of Sciences, University of Granada, Granada, Spain; Instituto de Investigación Biosanitaria ibs, Granada, Granada, Spain; Division of Obstetrics and Gynecology, Department of Clinical Science, Intervention and Technology (CLINTEC), Karolinska Institutet and Karolinska University Hospital, Stockholm, Sweden
| | - Josep Pla
- Reproductive Genetics Unit, IVIRMA Global, Barcelona, Spain.
| |
Collapse
|
3
|
Greco E, Greco PF, Listorti I, Ronsini C, Cucinelli F, Biricik A, Viotti M, Meschino N, Spinella F. The mosaic embryo: what it means for the doctor and the patient. Minerva Obstet Gynecol 2024; 76:89-101. [PMID: 37427860 DOI: 10.23736/s2724-606x.23.05281-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/11/2023]
Abstract
INTRODUCTION Mosaic embryos are embryos that on preimplantation genetic analysis are found to be composed of euploid and aneuploid cells. Although most of these embryos do not implant when transferred into the uterus following IVF treatment, some may implant and are capable of giving rise to babies. EVIDENCE ACQUISITION There is currently an increasing number of reports of live births following the transfer of mosaic embryos. Compared to euploid, mosaic embryos have lower implantation rates and higher rates of miscarriage, and occasionally aneuploid component persists. However, their outcome is better than that obtained after the transfer of embryos consisting entirely of aneuploid cells. After implantation, the ability to develop into a full-term pregnancy is influenced by the amount and type of chromosomal mosaicism present in a mosaic embryo. Nowadays many experts in the reproductive field consider mosaic transfers as an option when no euploid embryos are available. Genetic counseling is an important part of educating patients about the likelihood of having a pregnancy with healthy baby but also on the risk that mosaicism could persist and result in liveborn with chromosomal abnormality. Each situation needs to be assessed on a case-by-case basis and counseled accordingly. EVIDENCE SYNTHESIS So far, the transfers of 2155 mosaic embryos have been documented and 440 live births resulting in healthy babies have been reported. In addition, in the literature to date, there are 6 cases in which embryonic mosaicism persisted. CONCLUSIONS In conclusion, the available data indicate that mosaic embryos have the potential to implant and develop into healthy babies, albeit with lower success rates than euploids. Further clinical outcomes should be collected to better establish a refined ranking of embryos to transfer.
Collapse
Affiliation(s)
- Ermanno Greco
- Department of Obstetrics and Gynecology, UniCamillus International University, Rome, Italy
- Villa Mafalda, Centre For Reproductive Medicine, Rome, Italy
| | - Pier F Greco
- Villa Mafalda, Centre For Reproductive Medicine, Rome, Italy
| | - Ilaria Listorti
- Villa Mafalda, Centre For Reproductive Medicine, Rome, Italy
| | - Carlo Ronsini
- Department of Women and Children, Luigi Vanvitelli University of Campania, Naples, Italy
- Department of General and Specialist Surgery, Luigi Vanvitelli University of Campania, Naples, Italy
| | - Francesco Cucinelli
- Reproductive Unit, Department of Obstetrics and Gynaecology, San Camillo Forlanini Hospital, Rome, Italy
| | | | - Manuel Viotti
- Kindlabs, Kindbody, New York, NY, USA
- Zouves Foundation for Reproductive Medicine, Foster City, CA, USA
| | | | | |
Collapse
|
4
|
Popovic M, Borot L, Lorenzon AR, Lopes ALRDC, Sakkas D, Lledó B, Morales R, Ortiz JA, Polyzos NP, Parriego M, Azpiroz F, Galain M, Pujol A, Menten B, Dhaenens L, Vanden Meerschaut F, Stoop D, Rodriguez M, de la Blanca EP, Rodríguez A, Vassena R. Implicit bias in diagnosing mosaicism amongst preimplantation genetic testing providers: results from a multicenter study of 36 395 blastocysts. Hum Reprod 2024; 39:258-274. [PMID: 37873575 DOI: 10.1093/humrep/dead213] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Revised: 09/15/2023] [Indexed: 10/25/2023] Open
Abstract
STUDY QUESTION Does the diagnosis of mosaicism affect ploidy rates across different providers offering preimplantation genetic testing for aneuploidies (PGT-A)? SUMMARY ANSWER Our analysis of 36 395 blastocyst biopsies across eight genetic testing laboratories revealed that euploidy rates were significantly higher in providers reporting low rates of mosaicism. WHAT IS KNOWN ALREADY Diagnoses consistent with chromosomal mosaicism have emerged as a third category of possible embryo ploidy outcomes following PGT-A. However, in the era of mosaicism, embryo selection has become increasingly complex. Biological, technical, analytical, and clinical complexities in interpreting such results have led to substantial variability in mosaicism rates across PGT-A providers and clinics. Critically, it remains unknown whether these differences impact the number of euploid embryos available for transfer. Ultimately, this may significantly affect clinical outcomes, with important implications for PGT-A patients. STUDY DESIGN, SIZE, DURATION In this international, multicenter cohort study, we reviewed 36 395 consecutive PGT-A results, obtained from 10 035 patients across 11 867 treatment cycles, conducted between October 2015 and October 2021. A total of 17 IVF centers, across eight PGT-A providers, five countries and three continents participated in the study. All blastocysts were tested using trophectoderm biopsy and next-generation sequencing. Both autologous and donation cycles were assessed. Cycles using preimplantation genetic testing for structural rearrangements were excluded from the analysis. PARTICIPANTS/MATERIALS, SETTING, METHODS The PGT-A providers were randomly categorized (A to H). Providers B, C, D, E, F, G, and H all reported mosaicism, whereas Provider A reported embryos as either euploid or aneuploid. Ploidy rates were analyzed using multilevel mixed linear regression. Analyses were adjusted for maternal age, paternal age, oocyte source, number of embryos biopsied, day of biopsy, and PGT-A provider, as appropriate. We compared associations between genetic testing providers and PGT-A outcomes, including the number of chromosomally normal (euploid) embryos determined to be suitable for transfer. MAIN RESULTS AND THE ROLE OF CHANCE The mean maternal age (±SD) across all providers was 36.2 (±5.2). Our findings reveal a strong association between PGT-A provider and the diagnosis of euploidy and mosaicism. Amongst the seven providers that reported mosaicism, the rates varied from 3.1% to 25.0%. After adjusting for confounders, we observed a significant difference in the likelihood of diagnosing mosaicism across providers (P < 0.001), ranging from 6.5% (95% CI: 5.2-7.4%) for Provider B to 35.6% (95% CI: 32.6-38.7%) for Provider E. Notably, adjusted euploidy rates were highest for providers that reported the lowest rates of mosaicism (Provider B: euploidy, 55.7% (95% CI: 54.1-57.4%), mosaicism, 6.5% (95% CI: 5.2-7.4%); Provider H: euploidy, 44.5% (95% CI: 43.6-45.4%), mosaicism, 9.9% (95% CI: 9.2-10.6%)); and Provider D: euploidy, 43.8% (95% CI: 39.2-48.4%), mosaicism, 11.0% (95% CI: 7.5-14.5%)). Moreover, the overall chance of having at least one euploid blastocyst available for transfer was significantly higher when mosaicism was not reported, when we compared Provider A to all other providers (OR = 1.30, 95% CI: 1.13-1.50). Differences in diagnosing and interpreting mosaic results across PGT-A laboratories raise further concerns regarding the accuracy and relevance of mosaicism predictions. While we confirmed equivalent clinical outcomes following the transfer of mosaic and euploid blastocysts, we found that a significant proportion of mosaic embryos are not used for IVF treatment. LIMITATIONS, REASONS FOR CAUTION Due to the retrospective nature of the study, associations can be ascertained, however, causality cannot be established. Certain parameters such as blastocyst grade were not available in the dataset. Furthermore, certain platform-related and clinic-specific factors may not be readily quantifiable or explicitly captured in our dataset. As such, a full elucidation of all potential confounders accounting for variability may not be possible. WIDER IMPLICATIONS OF THE FINDINGS Our findings highlight the strong need for standardization and quality assurance in the industry. The decision not to transfer mosaic embryos may ultimately reduce the chance of success of a PGT-A cycle by limiting the pool of available embryos. Until we can be certain that mosaic diagnoses accurately reflect biological variability, reporting mosaicism warrants utmost caution. A prudent approach is imperative, as it may determine the difference between success or failure for some patients. STUDY FUNDING/COMPETING INTEREST(S) This work was supported by the Torres Quevedo Grant, awarded to M.P. (PTQ2019-010494) by the Spanish State Research Agency, Ministry of Science and Innovation, Spain. M.P., L.B., A.R.L., A.L.R.d.C.L., N.P.P., M.P., D.S., F.A., A.P., B.M., L.D., F.V.M., D.S., M.R., E.P.d.l.B., A.R., and R.V. have no competing interests to declare. B.L., R.M., and J.A.O. are full time employees of IB Biotech, the genetics company of the Instituto Bernabeu group, which performs preimplantation genetic testing. M.G. is a full time employee of Novagen, the genetics company of Cegyr, which performs preimplantation genetic testing. TRIAL REGISTRATION NUMBER N/A.
Collapse
Affiliation(s)
- Mina Popovic
- Research and Development, Eugin Group, Barcelona, Spain
| | - Lorena Borot
- Research and Development, Eugin Group, Barcelona, Spain
| | | | | | | | | | | | | | - Nikolaos P Polyzos
- Clínica Dexeus Mujer, Dexeus University Hospital, Barcelona, Spain
- Department of Reproductive Medicine, Ghent University Hospital, Ghent, Belgium
| | - Mónica Parriego
- Clínica Dexeus Mujer, Dexeus University Hospital, Barcelona, Spain
| | - Felicitas Azpiroz
- Research and Development, Eugin Group, Barcelona, Spain
- Cegyr-Medicina y Genética Reproductiva-Eugin Group, Buenos Aires, Argentina
| | - Micaela Galain
- Cegyr-Medicina y Genética Reproductiva-Eugin Group, Buenos Aires, Argentina
| | - Aïda Pujol
- Center for Infertility and Human Reproduction, CIRH-Eugin Group, Barcelona, Spain
| | - Björn Menten
- Center for Medical Genetics, Ghent University Hospital, Ghent, Belgium
| | - Lien Dhaenens
- Department of Reproductive Medicine, Ghent University Hospital, Ghent, Belgium
| | | | - Dominic Stoop
- Department of Reproductive Medicine, Ghent University Hospital, Ghent, Belgium
| | | | | | | | - Rita Vassena
- Research and Development, Eugin Group, Barcelona, Spain
| |
Collapse
|
5
|
Sanders KD, Griffin DK, Martell HJ, Blazek J, Large M, Gordon T. What proportion of embryos should be considered for transfer following a mosaic diagnosis? A study of 115 clinics from a central diagnostic laboratory. J Assist Reprod Genet 2023; 40:653-664. [PMID: 36708429 PMCID: PMC10033805 DOI: 10.1007/s10815-022-02678-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Accepted: 12/02/2022] [Indexed: 01/29/2023] Open
Abstract
PURPOSE The aim of this study is to identify what proportion of mosaic embryo diagnoses should be considered for transfer, and thereby assess the impact on patient cases. METHODS We categorised mosaic embryos into 3 groups; high, medium and low priority for transfer based on the percentage of biopsy sample being aneuploid and the chromosomes involved. The categories were applied to those patients that had no euploid embryo diagnoses but 1 or more mosaic embryos identified as mosaic available after PGT-A. RESULTS In total, 6614 PGT-A cases from 115 clinics and a single diagnostic laboratory were reviewed. Further, 1384 [20.9%] cases only had aneuploid embryos, 4538 [68.6%] cases had one or more euploid embryos and 692 [10.5%] cases had no euploid and one or more mosaic embryo. The mosaic embryos in the no euploid, one or more mosaic group, when reviewed using priorities, resulted in: 111 [1.7%] of cases having at least one high priority mosaic available. 184 [2.8%] of cases having no high priority but at least one medium priority mosaic available. 397 [6.0%] of cases only having low priority mosaic embryos available. CONCLUSION Based on this data, embryos identified as mosaic will only be considered for transfer in the first instance for around 4.5% (when taking high and medium priority and excluding low priority cases) of all PGT-A cases.
Collapse
Affiliation(s)
| | - Darren K Griffin
- School of Biosciences, University of Kent, Canterbury, CT2 7NJ, UK
| | - Henry J Martell
- School of Biosciences, University of Kent, Canterbury, CT2 7NJ, UK
| | | | | | | |
Collapse
|
6
|
Cheng L, Meiser B, Kennedy D, Kirk E, Barlow-Stewart K, Kaur R. Exploration of decision-making regarding the transfer of mosaic embryos following preimplantation genetic testing: a qualitative study. Hum Reprod Open 2022; 2022:hoac035. [PMID: 36157005 PMCID: PMC9492260 DOI: 10.1093/hropen/hoac035] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Revised: 08/10/2022] [Indexed: 11/24/2022] Open
Abstract
STUDY QUESTION What are patients’ reasoning and decisional needs in relation to the transfer of mosaic embryos following preimplantation genetic testing (PGT)? SUMMARY ANSWER This study identified four themes, which were patients’ reasoning behind decision-making, their decisional needs, the influence of the mosaic embryos on the decision-making and the role of health professionals. WHAT IS KNOWN ALREADY To date, no study has investigated the reasoning of patients behind their decision-making and the influence of mosaic embryos. STUDY DESIGN, SIZE, DURATION This is a cross-sectional study using a qualitative approach. Twenty participants were interviewed, and recruitment was ceased when no new information was identified in the data analysis. It ensured a sufficient sample size for a qualitative study. PARTICIPANTS/MATERIALS, SETTING, METHODS Participants were females with mosaic embryos. Semi-structured in-depth interviews were conducted via telephone. MAIN RESULTS AND THE ROLE OF CHANCE Four themes were identified: reasoning behind decision-making, decisional needs, influence of mosaic embryos on decision-making and the role of health professionals. Potential risks of transferring mosaic embryos and prioritization of euploid embryos were the main reasons for not transferring mosaic embryos. A lack of alternatives, perceived benefits and risk tolerance were main reasons for transferring mosaic embryos. Patients reported that information on mosaic embryos, amniocentesis and termination was important to support their decision-making. Unmet needs relating to healthcare services and social support were reported. In addition, having mosaic embryos affected the patients’ emotional and behavioural responses, discussions about prenatal testing, attitudes to termination and further IVF cycles and attitudes towards PGT. Health professionals were found to influence the patients’ decision-making. LIMITATIONS, REASONS FOR CAUTION Participants were recruited through one clinic, which may limit the transferability of results. Also, patients’ experiences in relation to financial aspects of PGT may not be relevant to other jurisdictions due to different healthcare policies. WIDER IMPLICATIONS OF THE FINDINGS The results may inform how clinicians provide healthcare services based on factors influencing patients’ decision-making. Health professionals should be aware of the influence their attitudes can have on patients’ decision-making and should present information accordingly. Also, providing all relevant information may help to facilitate informed decision-making. Provision of psychological support from professionals and support groups is also critical during the process of testing and transfer. Patients have educational needs regarding mosaic embryos, and educational resources including decision aids in plain language are needed. STUDY FUNDING/COMPETING INTEREST(S) B.M. was funded through a Senior Research Fellowship Level B (ID 1078523) from the National Health and Medical Research Council of Australia. L.C. was supported by a University International Postgraduate Award under the Australian Government Research Training Program (RTP) scholarship. No other funding was received for this study. The authors report no competing interests. TRIAL REGISTRATION NUMBER N/A.
Collapse
Affiliation(s)
- Lin Cheng
- University of New South Wales Prince of Wales Clinical School, , NSW, Australia. Level 4, Lowy Cancer Research Centre, UNSW Sydney, Randwick 2052, NSW, Australia
| | - Bettina Meiser
- University of New South Wales Prince of Wales Clinical School, , NSW, Australia. Level 4, Lowy Cancer Research Centre, UNSW Sydney, Randwick 2052, NSW, Australia
| | - Debra Kennedy
- Royal Hospital for Women; IVFAustralia, NSW , Australia. MotherSafe, Royal Hospital for Women, Barker St, Randwick 2052, NSW, Australia
- Royal Hospital for Women; IVFAustralia, NSW , Australia. IVFAustralia, Level1/33 York St, Sydney, NSW 2000, Australia
| | - Edwin Kirk
- University of New South Wales Sydney Children's Hospital; School of Women's and Children's Health, , NSW, Australia. Centre for Clinical Genetics, Bright Alliance Building, High Street, Randwick 2031, NSW, Australia
| | - Kristine Barlow-Stewart
- The University of Sydney Northern Clinical School, Faculty of Medicine and Health, , NSW, Australia. Northern Clinical School, Faculty of Medicine and Health, The University of Sydney, NSW, Australia
| | - Rajneesh Kaur
- The University of Sydney Faculty of Medicine and Health Administration, , NSW, Australia. Faculty of Medicine and Health Administration, Sydney Medical School's Education Office, The University of Sydney, NSW, Australia
| |
Collapse
|
7
|
Allais A, FitzHarris G. Absence of a robust mitotic timer mechanism in early preimplantation mouse embryos leads to chromosome instability. Development 2022; 149:275859. [DOI: 10.1242/dev.200391] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2021] [Accepted: 05/12/2022] [Indexed: 01/07/2023]
Abstract
ABSTRACT
Preimplantation embryos often consist of a combination of euploid and aneuploid cells, suggesting that safeguards preventing the generation and propagation of aneuploid cells in somatic cells might be deficient in embryos. In somatic cells, a mitotic timer mechanism has been described, in which even a small increase in the duration of M phase can cause a cell cycle arrest in the subsequent interphase, preventing further propagation of cells that have undergone a potentially hazardously long M phase. Here, we report that cell divisions in the mouse embryo and embryonic development continue even after a mitotic prolongation of several hours. However, similar M-phase extensions caused cohesion fatigue, resulting in prematurely separated sister chromatids and the production of micronuclei. Only extreme prolongation of M phase caused a subsequent interphase arrest, through a mechanism involving DNA damage. Our data suggest that the simultaneous absence of a robust mitotic timer and susceptibility of the embryo to cohesion fatigue could contribute to chromosome instability in mammalian embryos.
This article has an associated ‘The people behind the papers’ interview.
Collapse
Affiliation(s)
- Adélaïde Allais
- Centre de recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM) 1 , H2X 0A9 Montréal, Québec , Canada
| | - Greg FitzHarris
- Centre de recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM) 1 , H2X 0A9 Montréal, Québec , Canada
- Université de Montréal 2 Department of OBGYN, and Department of Pathology and Cell Biology , , H3T 1C5 Montréal, Québec , Canada
| |
Collapse
|
8
|
Regin M, Spits C, Sermon K. On the origins and fate of chromosomal abnormalities in human preimplantation embryos: an unsolved riddle. Mol Hum Reprod 2022; 28:6566308. [DOI: 10.1093/molehr/gaac011] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 03/30/2022] [Indexed: 11/13/2022] Open
Abstract
Abstract
About 8 out of 10 human embryos obtained in vitro harbour chromosomal abnormalities of either meiotic or mitotic origin. Abnormalities of mitotic origin lead to chromosomal mosaicism, a phenomenon which has sparked much debate lately as it confounds results obtained through preimplantation genetic testing for aneuploidy (PGT-A). PGT-A in itself is still highly debated, not only on the modalities of its execution, but also on whether it should be offered to patients at all.
We will focus on post-zygotic chromosomal abnormalities leading to mosaicism. First, we will summarize what is known of the rates of chromosomal abnormalities at different developmental stages. Next, based on the current understanding of the origin and cellular consequences of chromosomal abnormalities, which is largely based on studies on cancer cells and model organisms, we will offer a number of hypotheses on which mechanisms may be at work in early human development. Finally, and very briefly, we will touch upon the impact our current knowledge has on the practice of PGT-A. What is the level of abnormal cells that an embryo can tolerate before it loses its potential for full development? And is blastocyst biopsy as harmless as it seems?
Collapse
Affiliation(s)
- Marius Regin
- Research group Reproduction and Genetics, Vrije Universiteit Brussel, Brussels, 1090, Belgium
| | - Claudia Spits
- Research group Reproduction and Genetics, Vrije Universiteit Brussel, Brussels, 1090, Belgium
| | - Karen Sermon
- Research group Reproduction and Genetics, Vrije Universiteit Brussel, Brussels, 1090, Belgium
| |
Collapse
|
9
|
Ai X, Shi Y, Liu LW, Xu Y, Zhang H, Liu Y, Wang J, Ding C, Cai B, Zhou C, Xu Y. Risk factors related to chromosomal mosaicism in human blastocysts. Reprod Biomed Online 2022; 45:54-62. [DOI: 10.1016/j.rbmo.2022.02.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Revised: 02/16/2022] [Accepted: 02/22/2022] [Indexed: 11/24/2022]
|
10
|
Zadesenets KS, Rubtsov NB. B Chromosomes in Free-Living Flatworms of the Genus Macrostomum (Platyhelminthes, Macrostomorpha). Int J Mol Sci 2021; 22:13617. [PMID: 34948412 PMCID: PMC8708343 DOI: 10.3390/ijms222413617] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Revised: 12/14/2021] [Accepted: 12/17/2021] [Indexed: 11/17/2022] Open
Abstract
B chromosomes (Bs) or supernumerary chromosomes are extra chromosomes in the species karyotype that can vary in its copy number. Bs are widespread in eukaryotes. Usually, the Bs of specimens collected from natural populations are the object of the B chromosome studies. We applied another approach analyzing the Bs in animals maintained under the laboratory conditions as lines and cultures. In this study, three species of the Macrostomum genus that underwent a recent whole-genome duplication (WGD) were involved. In laboratory lines of M. lignano and M. janickei, the frequency of Bs was less than 1%, while in the laboratory culture of M. mirumnovem, it was nearer 30%. Their number in specimens of the culture varied from 1 to 14. Mosaicism on Bs was discovered in parts of these animals. We analyzed the distribution of Bs among the worms of the laboratory cultures during long-term cultivation, the transmission rates of Bs in the progeny obtained from crosses of worms with different numbers of Bs, and from self-fertilized isolated worms. The DNA content of the Bs in M. mirumnovem was analyzed with the chromosomal in situ suppression (CISS) hybridization of microdissected DNA probes derived from A chromosomes (As). Bs mainly consisted of repetitive DNA. The cytogenetic analysis also revealed the divergence and high variation in large metacentric chromosomes (LMs) containing numerous regions enriched for repeats. The possible mechanisms of the appearance and evolution of Bs and LMs in species of the Macrostomum genus were also discussed.
Collapse
Affiliation(s)
- Kira S. Zadesenets
- The Federal Research Center Institute of Cytology and Genetics SB RAS, 630090 Novosibirsk, Russia;
| | | |
Collapse
|
11
|
Mastenbroek S, de Wert G, Adashi EY. The Imperative of Responsible Innovation in Reproductive Medicine. N Engl J Med 2021; 385:2096-2100. [PMID: 34818487 DOI: 10.1056/nejmsb2101718] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Affiliation(s)
- Sebastiaan Mastenbroek
- From the Center for Reproductive Medicine, Amsterdam Reproduction and Development Research Institute, Amsterdam UMC, University of Amsterdam, Amsterdam (S.M.), and the Department of Health, Ethics and Society, Research School GROW, Maastricht University, Maastricht (G.W.) - both in the Netherlands; and the Department of Medical Science, Brown University, Providence, RI (E.Y.A.)
| | - Guido de Wert
- From the Center for Reproductive Medicine, Amsterdam Reproduction and Development Research Institute, Amsterdam UMC, University of Amsterdam, Amsterdam (S.M.), and the Department of Health, Ethics and Society, Research School GROW, Maastricht University, Maastricht (G.W.) - both in the Netherlands; and the Department of Medical Science, Brown University, Providence, RI (E.Y.A.)
| | - Eli Y Adashi
- From the Center for Reproductive Medicine, Amsterdam Reproduction and Development Research Institute, Amsterdam UMC, University of Amsterdam, Amsterdam (S.M.), and the Department of Health, Ethics and Society, Research School GROW, Maastricht University, Maastricht (G.W.) - both in the Netherlands; and the Department of Medical Science, Brown University, Providence, RI (E.Y.A.)
| |
Collapse
|
12
|
Zheng W, Yang C, Yang S, Sun S, Mu M, Rao M, Zu R, Yan J, Ren B, Yang R, Guan Y. Obstetric and neonatal outcomes of pregnancies resulting from preimplantation genetic testing: a systematic review and meta-analysis. Hum Reprod Update 2021; 27:989-1012. [PMID: 34473268 DOI: 10.1093/humupd/dmab027] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Revised: 07/10/2021] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Preimplantation genetic testing (PGT) includes methods that allow embryos to be tested for severe inherited diseases or chromosomal abnormalities. In addition to IVF/ICSI and repeated freezing and thawing of the embryos, PGT requires a biopsy to obtain embryonic genetic material for analysis. However, the potential effects of PGT on obstetric and neonatal outcomes are currently uncertain. OBJECTIVE AND RATIONALE This study aimed to investigate whether pregnancies conceived after PGT were associated with a higher risk of adverse obstetric and neonatal outcomes compared with spontaneously conceived (SC) pregnancies or pregnancies conceived after IVF/ICSI. SEARCH METHODS PubMed, EMBASE, MEDLINE, Web of Science and The Cochrane Library entries from January 1990 to January 2021 were searched. The primary outcomes in this study were low birth weight (LBW) and congenital malformations (CMs), and the secondary outcomes included gestational age, preterm delivery (PTD), very preterm delivery (VPTD), birth weight (BW), very low birth weight (VLBW), neonatal intensive care unit (NICU) admission, hypertensive disorders of pregnancy (HDP), gestational diabetes, placenta previa and preterm premature rupture of membranes (PROM). We further pooled the results of PGT singleton pregnancies. Subgroup analyses included preimplantation genetic diagnosis (PGD), preimplantation genetic screening (PGS), cleavage-stage biopsy combined with fresh embryo transfer (CB-ET) and blastocyst biopsy combined with frozen-thawed embryo transfer (BB-FET). OUTCOMES This meta-analysis included 15 studies involving 3682 babies born from PGT pregnancies, 127 719 babies born from IVF/ICSI pregnancies and 915 222 babies born from SC pregnancies. The relative risk (RR) of LBW was higher in PGT pregnancies compared with SC pregnancies (RR = 3.95, 95% confidence interval [CI]: 2.32-6.72), but the risk of CMs was not different between the two groups. The pooled results for the risks of LBW and CMs were similar in PGT and IVF/ICSI pregnancies. The risks of PTD (RR = 3.12, 95% CI: 2.67-3.64) and HDP (RR = 3.12, 95% CI: 2.18-4.47) were significantly higher in PGT pregnancies compared with SC pregnancies. Lower gestational age (mean difference [MD] = -0.76 weeks, 95% CI -1.17 to -0.34) and BW (MD = -163.80 g, 95% CI: -299.35 to -28.24) were also noted for PGT pregnancies compared with SC pregnancies. Nevertheless, compared with IVF/ICSI pregnancies, the risks of VPTD and VLBW in PGT pregnancies were significantly decreased by 41% and 30%, respectively, although the risk of HDP was still significantly increased by 50% in PGT pregnancies compared with IVF/ICSI pregnancies. The combined results of obstetric and neonatal outcomes of PGT and IVF/ICSI singleton pregnancies were consistent with the overall results. Further subgroup analyses indicated that both PGD and PGS pregnancies were associated with a higher risk of PTD and a lower gestational age compared with SC pregnancies. WIDER IMPLICATIONS This meta-analysis showed that PGT pregnancies may be associated with increased risks of LBW, PTD and HDP compared with SC pregnancies. The overall obstetric and neonatal outcomes of PGT pregnancies are favourable compared with those of IVF/ICSI pregnancies, although PGT pregnancies were associated with a higher risk of HDP. However, because the number of studies that could be included was limited, more randomised controlled trials and prospective cohort studies are needed to confirm these conclusions.
Collapse
Affiliation(s)
- Wei Zheng
- Center for Reproductive Medicine, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Chen Yang
- Center for Reproductive Medicine, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Shuheng Yang
- Center for Reproductive Medicine, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Simin Sun
- Center for Reproductive Medicine, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Mingkun Mu
- Center for Reproductive Medicine, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Meng Rao
- Department of Reproduction and Genetics, The First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Ruowen Zu
- Center for Reproductive Medicine, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Junfang Yan
- Center for Reproductive Medicine, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Bingnan Ren
- Center for Reproductive Medicine, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Rujing Yang
- Center for Reproductive Medicine, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yichun Guan
- Center for Reproductive Medicine, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
13
|
Viotti M, McCoy RC, Griffin DK, Spinella F, Greco E, Madjunkov M, Madjunkova S, Librach CL, Victor AR, Barnes FL, Zouves CG. Let the data do the talking: the need to consider mosaicism during embryo selection. Fertil Steril 2021; 116:1212-1219. [PMID: 34627598 DOI: 10.1016/j.fertnstert.2021.09.008] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2021] [Revised: 08/27/2021] [Accepted: 09/08/2021] [Indexed: 01/01/2023]
Abstract
Chromosomal mosaicism, the coexistence of cells with different chromosomal content, has been documented in human embryos for 3 decades. Early versions of preimplantation genetic testing for aneuploidy (PGT-A) did not measure mosaicism, either because typically only a single cell was assessed or because the technique could not accurately identify it. Although this led to a straightforward diagnosis (an embryo was considered either normal or abnormal), it simply avoided the issue and, in hindsight, may have led to numerous misdiagnoses with negative clinical consequences. Modern PGT-A evaluates a multicellular biopsy specimen with techniques capable of recognizing intermediate copy number signals for chromosomes or subchromosomal regions. We are, therefore, inevitably confronted with the issue of mosaicism and the challenge of managing embryos producing such results in the clinic. Here we discuss recent data showing that not only mosaicism in general, but specific features of mosaicism detected with PGT-A, are associated with variable clinical outcomes. The conclusion is evident: mosaicism should be considered for more informed and improved embryo selection in the clinic.
Collapse
Affiliation(s)
- Manuel Viotti
- Zouves Foundation for Reproductive Medicine, Foster City, California; Zouves Fertility Center, Foster City, California.
| | - Rajiv C McCoy
- Department of Biology, Johns Hopkins University, Baltimore, Maryland
| | - Darren K Griffin
- School of Biosciences, University of Kent, Canterbury, United Kingdom
| | | | - Ermanno Greco
- Center for Reproductive Medicine, Villa Mafalda, Rome, Italy; Department of Obstetrics and Gynecology, UniCamillus International Medical University, Rome, Italy
| | - Mitko Madjunkov
- CReATe Fertility Centre, Toronto, Ontario, Canada; Department of Obstetrics and Gynecology, University of Toronto, Toronto, Ontario, Canada
| | - Svetlana Madjunkova
- CReATe Fertility Centre, Toronto, Ontario, Canada; Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
| | - Clifford L Librach
- CReATe Fertility Centre, Toronto, Ontario, Canada; Department of Obstetrics and Gynecology, University of Toronto, Toronto, Ontario, Canada; Institute of Medical Sciences and Department of Physiology, University of Toronto, Toronto, Ontario, Canada
| | - Andrea R Victor
- Zouves Fertility Center, Foster City, California; School of Biosciences, University of Kent, Canterbury, United Kingdom
| | - 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
| |
Collapse
|
14
|
Tang M, Popovic M, Stamatiadis P, Van der Jeught M, Van Coster R, Deforce D, De Sutter P, Coucke P, Menten B, Stoop D, Boel A, Heindryckx B. Germline nuclear transfer in mice may rescue poor embryo development associated with advanced maternal age and early embryo arrest. Hum Reprod 2021; 35:1562-1577. [PMID: 32613230 DOI: 10.1093/humrep/deaa112] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2019] [Revised: 04/06/2020] [Indexed: 01/06/2023] Open
Abstract
STUDY QUESTION Can pronuclear transfer (PNT) or maternal spindle transfer (ST) be applied to overcome poor embryo development associated with advanced maternal age or early embryo arrest in a mouse model? SUMMARY ANSWER Both PNT and ST may have the potential to restore embryonic developmental potential in a mouse model of reproductive ageing and embryonic developmental arrest. WHAT IS KNOWN ALREADY Germline nuclear transfer (NT) techniques, such as PNT and ST, are currently being applied in humans to prevent the transmission of mitochondrial diseases. Yet, there is also growing interest in the translational use of NT for treating infertility and improving IVF outcomes. Nevertheless, direct scientific evidence to support such applications is currently lacking. Moreover, it remains unclear which infertility indications may benefit from these novel assisted reproductive technologies. STUDY DESIGN, SIZE, DURATION We applied two mouse models to investigate the potential of germline NT for overcoming infertility. Firstly, we used a model of female reproductive ageing (B6D2F1 mice, n = 155), with ages ranging from 6 to 8 weeks (young), 56 (aged) to 70 weeks (very-aged), corresponding to a maternal age of <30, ∼36 and ∼45 years in humans, respectively. Secondly, we used NZB/OlaHsd female mice (7-14 weeks, n = 107), as a model of early embryo arrest. This mouse strain exhibits a high degree of two-cell block. Metaphase II (MII) oocytes and zygotes were retrieved following superovulation. PARTICIPANTS/MATERIALS, SETTING, METHODS Ovarian reserve was assessed by histological analysis in the reproductive-aged mice. Mitochondrial membrane potential (△Ψm) was measured by JC-1 staining in MII oocytes, while spindle-chromosomal morphology was examined by confocal microscopy. Reciprocal ST and PNT were performed by transferring the meiotic spindle or pronuclei (PN) from unfertilised or fertilised oocytes (after ICSI) to enucleated oocytes or zygotes between aged or very-aged and young mice. Similarly, NT was also conducted between NZB/OlaHsd (embryo arrest) and B6D2F1 (non-arrest control) mice. Finally, the effect of cytoplasmic transfer (CT) was examined by injecting a small volume (∼5%) of cytoplasm from the oocytes/zygotes of young (B6D2F1) mice to the oocytes/zygotes of aged or very-aged mice or embryo-arrest mice. Overall, embryonic developmental rates of the reconstituted PNT (n = 572), ST (n = 633) and CT (n = 336) embryos were assessed to evaluate the efficiency of these techniques. Finally, chromosomal profiles of individual NT-generated blastocysts were evaluated using next generation sequencing. MAIN RESULTS AND THE ROLE OF CHANCE Compared to young mice, the ovarian reserve in aged and very-aged mice was severely diminished, reflected by a lower number of ovarian follicles and a reduced number of ovulated oocytes (P < 0.001). Furthermore, we reveal that the average △Ψm in both aged and very-aged mouse oocytes was significantly reduced compared to young mouse oocytes (P < 0.001). In contrast, the average △Ψm in ST-reconstructed oocytes (very-aged spindle and young cytoplast) was improved in comparison to very-aged mouse oocytes (P < 0.001). In addition, MII oocytes from aged and very-aged mice exhibited a higher rate of abnormalities in spindle assembly (P < 0.05), and significantly lower fertilisation (60.7% and 45.3%) and blastocyst formation rates (51.4% and 38.5%) following ICSI compared to young mouse oocytes (89.7% and 87.3%) (P < 0.001). Remarkably, PNT from zygotes obtained from aged or very-aged mice to young counterparts significantly improved blastocyst formation rates (74.6% and 69.2%, respectively) (P < 0.05). Similarly, both fertilisation and blastocyst rates were significantly increased after ST between aged and young mice followed by ICSI (P < 0.05). However, we observed no improvement in embryo development rates when performing ST from very-aged to young mouse oocytes following ICSI (P > 0.05). In the second series of experiments, we primarily confirmed that the majority (61.8%) of in vivo zygotes obtained from NZB/OlaHsd mice displayed two-cell block during in vitro culture, coinciding with a significantly reduced blastocyst formation rate compared to the B6D2F1 mice (13.5% vs. 90.7%; P < 0.001). Notably, following the transfer of PN from the embryo-arrest (NZB/OlaHsd) zygotes to enucleated non-arrest (B6D2F1) counterparts, most reconstructed zygotes developed beyond the two-cell stage, leading to a significantly increased blastocyst formation rate (89.7%) (P < 0.001). Similar findings were obtained after implementing ST between NZB/OlaHsd and B6D2F1 mice, followed by ICSI. Conversely, the use of CT did not improve embryo development in reproductive-age mice nor in the embryo-arrest mouse model (P > 0.05). Surprisingly, chromosomal analysis revealed that euploidy rates in PNT and ST blastocysts generated following the transfer of very-aged PN to young cytoplasts and very-aged spindles to young cytoplasts were comparable to ICSI controls (with young mouse oocytes). A high euploidy rate was also observed in the blastocysts obtained from either PNT or ST between young mice. Conversely, the transfer of young PN and young spindles into very-aged cytoplasts led to a higher rate of chromosomal abnormalities in both PNT and ST blastocysts. LARGE SCALE DATA N/A. LIMITATIONS, REASONS FOR CAUTION The limited number of blastocysts analysed warrants careful interpretation. Furthermore, our observations should be cautiously extrapolated to humans given the inherent differences between mice and women in regards to various biological processes, including centrosome inheritance. The findings suggest that ST or PNT procedures may be able to avoid aneuploidies generated during embryo development, but they are not likely to correct aneuploidies already present in some aged MII oocytes. WIDER IMPLICATIONS OF THE FINDINGS To our knowledge, this is the first study to evaluate the potential of PNT and ST in the context of advanced maternal age and embryonic developmental arrest in a mouse model. Our data suggest that PNT, and to a lesser extent ST, may represent a novel reproductive strategy to restore embryo development for these indications. STUDY FUNDING/COMPETING INTEREST(S) M.T. is supported by grants from the China Scholarship Council (CSC) (Grant no. 201506160059) and the Special Research Fund from Ghent University (Bijzonder Onderzoeksfonds, BOF) (Grant no. 01SC2916 and no. 01SC9518). This research is also supported by the FWO-Vlaanderen (Flemish fund for scientific research, Grant no. G051017N, G051516N and G1507816N). The authors declare no competing interests. TRIAL REGISTRATION NUMBER N/A.
Collapse
Affiliation(s)
- M Tang
- Ghent-Fertility and Stem cell Team (G-FaST), Department for Reproductive Medicine, Ghent University Hospital, Ghent 9000, Belgium
| | - M Popovic
- Ghent-Fertility and Stem cell Team (G-FaST), Department for Reproductive Medicine, Ghent University Hospital, Ghent 9000, Belgium
| | - P Stamatiadis
- Ghent-Fertility and Stem cell Team (G-FaST), Department for Reproductive Medicine, Ghent University Hospital, Ghent 9000, Belgium
| | - M Van der Jeught
- Ghent-Fertility and Stem cell Team (G-FaST), Department for Reproductive Medicine, Ghent University Hospital, Ghent 9000, Belgium
| | - R Van Coster
- Department of Pediatric Neurology and Metabolism, Ghent University Hospital, Ghent 9000, Belgium
| | - D Deforce
- Laboratory of Pharmaceutical Biotechnology, Faculty of Pharmaceutical Sciences, Ghent University, Ghent 9000, Belgium
| | - P De Sutter
- Ghent-Fertility and Stem cell Team (G-FaST), Department for Reproductive Medicine, Ghent University Hospital, Ghent 9000, Belgium
| | - P Coucke
- Center for Medical Genetics Ghent (CMGG), Department of Biomolecular Medicine, Ghent University Hospital, Ghent 9000, Belgium
| | - B Menten
- Center for Medical Genetics Ghent (CMGG), Department of Biomolecular Medicine, Ghent University Hospital, Ghent 9000, Belgium
| | - D Stoop
- Ghent-Fertility and Stem cell Team (G-FaST), Department for Reproductive Medicine, Ghent University Hospital, Ghent 9000, Belgium
| | - A Boel
- Ghent-Fertility and Stem cell Team (G-FaST), Department for Reproductive Medicine, Ghent University Hospital, Ghent 9000, Belgium
| | - B Heindryckx
- Ghent-Fertility and Stem cell Team (G-FaST), Department for Reproductive Medicine, Ghent University Hospital, Ghent 9000, Belgium
| |
Collapse
|
15
|
Depletion of aneuploid cells in human embryos and gastruloids. Nat Cell Biol 2021; 23:314-321. [PMID: 33837289 DOI: 10.1038/s41556-021-00660-7] [Citation(s) in RCA: 96] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Accepted: 03/04/2021] [Indexed: 02/01/2023]
Abstract
Chromosomal instability leading to aneuploidy is pervasive in early human embryos1-3 and is considered as a major cause of infertility and pregnancy wastage4,5. Here we provide several lines of evidence that blastocysts containing aneuploid cells are worthy of in vitro fertilization transfer. First, we show clinically that aneuploid embryos can lead to healthy births, suggesting the presence of an in vivo mechanism to eliminate aneuploidy. Second, early development and cell specification modelled in micropatterned human 'gastruloids' grown in confined geometry show that aneuploid cells are depleted from embryonic germ layers, but not from extraembryonic tissue, by apoptosis in a bone morphogenetic protein 4 (BMP4)-dependent manner. Third, a small percentage of euploid cells rescues embryonic tissue in mosaic gastruloids when mixed with aneuploid cells. Finally, single-cell RNA-sequencing analysis of early human embryos revealed a decline of aneuploidy beginning on day 3. Our findings challenge two current dogmas: that a single trophectoderm biopsy at blastocyst stage to perform prenatal genetic testing can accurately determine the chromosomal make-up of a human embryo, and that aneuploid embryos should be withheld from embryo transfer in association with in vitro fertilization.
Collapse
|
16
|
Cimadomo D, Rienzi L, Capalbo A, Rubio C, Innocenti F, García-Pascual CM, Ubaldi FM, Handyside A. The dawn of the future: 30 years from the first biopsy of a human embryo. The detailed history of an ongoing revolution. Hum Reprod Update 2020; 26:453-473. [PMID: 32441746 DOI: 10.1093/humupd/dmaa019] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2019] [Revised: 03/25/2020] [Indexed: 01/20/2023] Open
Abstract
Following early studies showing no adverse effects, cleavage stage biopsy by zona drilling using acid Tyrode's solution, and removal of single blastomeres for preimplantation genetic testing (PGT) and identification of sex in couples at risk of X-linked disease, was performed by Handyside and colleagues in late 1989, and pregnancies reported in 1990. This method was later used for specific diagnosis of monogenic conditions, and a few years later also for chromosomal structural and/or numerical impairments, thereby establishing a valuable alternative option to prenatal diagnosis. This revolutionary approach in clinical embryology spread worldwide, and several other embryo biopsy strategies developed over three decades in a process that is still ongoing. The rationale of this narrative review is to outline the different biopsy approaches implemented across the years in the workflow of the IVF clinics that provided PGT: their establishment, the first clinical experiences, their downsides, evolution, improvement and standardization. The history ends with a glimpse of the future: minimally/non-invasive PGT and experimental embryo micromanipulation protocols. This grand theme review outlines a timeline of the evolution of embryo biopsy protocols, whose implementation is increasing worldwide together with the increasing application of PGT techniques in IVF. It represents a vade mecum especially for the past, present and upcoming operators and experts in this field to (re)live this history from its dawn to its most likely future.
Collapse
Affiliation(s)
- Danilo Cimadomo
- Clinica Valle Giulia, Genera Center for Reproductive Medicine, Rome, Italy
| | - Laura Rienzi
- Clinica Valle Giulia, Genera Center for Reproductive Medicine, Rome, Italy
| | - Antonio Capalbo
- Igenomix Italy, Marostica, Italy.,Dipartimento di Scienze Anatomiche, Istologiche, Medico Legali e dell'Apparato Locomotore, Sezione Istologia ed Embriologia Medica, University of Rome 'Sapienza', Rome, Italy
| | - Carmen Rubio
- R&D Department, Igenomix and Incliva, Valencia, Spain
| | - Federica Innocenti
- Clinica Valle Giulia, Genera Center for Reproductive Medicine, Rome, Italy
| | | | | | - Alan Handyside
- School of Biosciences, University of Kent, Canterbury, UK
| |
Collapse
|
17
|
Abstract
Despite the ever-increasing number of patients undergoing fertility treatments and the expanded use of genetic testing in this context, there has been limited focus in the literature on the involvement of genetics professionals in the assisted reproductive technology (ART) setting. Here we discuss the importance of genetic counseling within reproductive medicine. We review how genetic testing of embryos is performed, the process of gamete donation, the challenges associated with genetic testing, and the complexities of genetic test result interpretation.
Collapse
Affiliation(s)
- Debra Lilienthal
- The Ronald O. Perelman and Claudia Cohen Center for Reproductive Medicine, Weill Cornell Medicine, New York, New York 10021, USA
| | - Michelle Cahr
- California Cryobank Life Sciences, Los Angeles, California 90025, USA
| |
Collapse
|
18
|
Orvieto R, Shimon C, Rienstein S, Jonish-Grossman A, Shani H, Aizer A. Do human embryos have the ability of self-correction? Reprod Biol Endocrinol 2020; 18:98. [PMID: 33023576 PMCID: PMC7539487 DOI: 10.1186/s12958-020-00650-8] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Accepted: 09/09/2020] [Indexed: 12/20/2022] Open
Abstract
Human embryogenesis frequently coinciding with cell division mistakes contributing to pervasive embryonic aneuploidy/mosaicism. While embryo self-correction was elegantly demonstrated in mouse models, human studies are lacking. Here we are witness to human embryos ability to eliminate/expel abnormal blastomeres as cell debris/fragments. Each blastocyst and its corresponding debris were separated and underwent whole genome amplification. Seven of the 11 pairs of blastocysts and their corresponding cell debris/fragments revealed discordant results. Of the 9 euploid blastocysts, four showed euploid debris, while in the others, the debris were aneuploid. In the remaining pairs, the debris showed additional aneuploidy to those presented by their corresponding blastocyst. The observed ability of human embryos to self-correction doubts many invasive and non-invasive preimplantation testing for aneuploidy at the blastocyst stage, rendering high rate of false positive (discarding "good" embryos) by identifying the cell-free DNA originated from the expelled cell debris, as aneuploidy/mosaic blastocyst.
Collapse
Affiliation(s)
- Raoul Orvieto
- grid.413795.d0000 0001 2107 2845Infertility and IVF Unit, Department of Obstetrics and Gynecology, Chaim Sheba Medical Center (Tel Hashomer), 56261 Ramat Gan, Israel
- grid.12136.370000 0004 1937 0546Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
- grid.12136.370000 0004 1937 0546The Tarnesby-Tarnowski Chair for Family Planning and Fertility Regulation, Sackler Faculty of Medicine, Tel-Aviv University, Tel Aviv, Israel
| | - Chen Shimon
- grid.413795.d0000 0001 2107 2845Infertility and IVF Unit, Department of Obstetrics and Gynecology, Chaim Sheba Medical Center (Tel Hashomer), 56261 Ramat Gan, Israel
| | - Shlomit Rienstein
- grid.12136.370000 0004 1937 0546Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
- grid.413795.d0000 0001 2107 2845Danek Gertner Institute of Human Genetics, Sheba Medical Center, 56261 Ramat-Gan, Israel
| | - Anat Jonish-Grossman
- grid.413795.d0000 0001 2107 2845Danek Gertner Institute of Human Genetics, Sheba Medical Center, 56261 Ramat-Gan, Israel
| | - Hagit Shani
- grid.12136.370000 0004 1937 0546Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
- grid.413795.d0000 0001 2107 2845Danek Gertner Institute of Human Genetics, Sheba Medical Center, 56261 Ramat-Gan, Israel
| | - Adva Aizer
- grid.413795.d0000 0001 2107 2845Infertility and IVF Unit, Department of Obstetrics and Gynecology, Chaim Sheba Medical Center (Tel Hashomer), 56261 Ramat Gan, Israel
- grid.12136.370000 0004 1937 0546Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| |
Collapse
|
19
|
Coll L, Parriego M, Mateo S, García-Monclús S, Rodríguez I, Boada M, Coroleu B, Polyzos NP, Vidal F, Veiga A. Prevalence, types and possible factors influencing mosaicism in IVF blastocysts: results from a single setting. Reprod Biomed Online 2020; 42:55-65. [PMID: 33153932 DOI: 10.1016/j.rbmo.2020.09.025] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Revised: 09/08/2020] [Accepted: 09/29/2020] [Indexed: 12/11/2022]
Abstract
RESEARCH QUESTION Are intrinsic or extrinsic factors associated with embryo mosaicism prevalence in IVF cycles? DESIGN Retrospective cohort study of preimplantation genetic testing for aneuploidy (PGT-A) cycles carried out at a university-affiliated IVF clinic between October 2017 and October 2019. Trophectoderm biopsies were analysed by next generation sequencing. Mosaicism prevalence, type of anomaly and the chromosomes involved were analysed. Intrinsic and extrinsic factors potentially inducing mosaicism were studied: maternal and paternal age, antral follicle count, cumulus-oocyte complexes retrieved, female body mass index, PGT-A indication, sperm concentration, total dosage of gonadotrophins, embryo quality and day of blastocyst formation, single-step commercial media used and biopsy operator. RESULTS Overall prevalence of mosaicism in our PGT-A setting was 13.9%. In segmental mosaicism, larger chromosomes tended to be more affected, which was not observed in whole-chromosome mosaicism. Additionally, segmental mosaicism was mostly observed in monosomy (69.6%; P < 0.01) compared with whole-chromosome mosaicism (49.7% monosomies versus 50.3% trisomies; P = 0.83). Although a high inter-patient variability was observed, only paternal age showed a positive association with mosaicism (adjusted OR 1.26, 95% CI 1.02 to 1.54) among the analysed variables. CONCLUSIONS Our results suggest remarkable differences in the mechanisms generating segmental and whole-chromosome mosaicism, indicating that they may deserve different consideration when studying them and when prioritizing them for transfer. Male factor seems to be associated with mosaicism and may be worthy of specific assessment in future studies.
Collapse
Affiliation(s)
- Lluc Coll
- Reproductive Medicine Unit, Department of Obstetrics, Gynaecology and Reproduction, Dexeus Mujer, Hospital Universitari Dexeus, Gran Via Carles III, 71-75, Barcelona 08028, Spain.
| | - Mònica Parriego
- Reproductive Medicine Unit, Department of Obstetrics, Gynaecology and Reproduction, Dexeus Mujer, Hospital Universitari Dexeus, Gran Via Carles III, 71-75, Barcelona 08028, Spain
| | - Sílvia Mateo
- Reproductive Medicine Unit, Department of Obstetrics, Gynaecology and Reproduction, Dexeus Mujer, Hospital Universitari Dexeus, Gran Via Carles III, 71-75, Barcelona 08028, Spain
| | - Sílvia García-Monclús
- Reproductive Medicine Unit, Department of Obstetrics, Gynaecology and Reproduction, Dexeus Mujer, Hospital Universitari Dexeus, Gran Via Carles III, 71-75, Barcelona 08028, Spain
| | - Ignacio Rodríguez
- Reproductive Medicine Unit, Department of Obstetrics, Gynaecology and Reproduction, Dexeus Mujer, Hospital Universitari Dexeus, Gran Via Carles III, 71-75, Barcelona 08028, Spain
| | - Montserrat Boada
- Reproductive Medicine Unit, Department of Obstetrics, Gynaecology and Reproduction, Dexeus Mujer, Hospital Universitari Dexeus, Gran Via Carles III, 71-75, Barcelona 08028, Spain
| | - Buenaventura Coroleu
- Reproductive Medicine Unit, Department of Obstetrics, Gynaecology and Reproduction, Dexeus Mujer, Hospital Universitari Dexeus, Gran Via Carles III, 71-75, Barcelona 08028, Spain
| | - Nikolaos P Polyzos
- Reproductive Medicine Unit, Department of Obstetrics, Gynaecology and Reproduction, Dexeus Mujer, Hospital Universitari Dexeus, Gran Via Carles III, 71-75, Barcelona 08028, Spain
| | - Francesca Vidal
- Unitat de Biologia Cellular, Facultat de Biociències, Universitat Autònoma de Barcelona, Bellaterra 08193, Spain
| | - Anna Veiga
- Reproductive Medicine Unit, Department of Obstetrics, Gynaecology and Reproduction, Dexeus Mujer, Hospital Universitari Dexeus, Gran Via Carles III, 71-75, Barcelona 08028, Spain; Barcelona Stem Cell Bank, IDIBELL Programme for Regenerative Medicine, 08908 L'Hospitalet de Llobregat, Spain
| |
Collapse
|
20
|
Nigmatova N, Abdilmanova B, Schigolev V. THE REPORTED SUCCESSFUL BABY DELIVERY AFTER PREIMPLANTATION GENETIC TESTING FOR ANEUPLOIDIES (PGT-A) BY MEANS OF NEXT GENERATION SEQUENCING (NGS). REPRODUCTIVE MEDICINE 2020. [DOI: 10.37800/rm2020-1-10] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The aim of this case report is to demonstrate a successful delivery of a baby after transfer of a blastocyst tested for aneuplodies by means of NGS. A woman aged 35 having two miscarriages decided for ICSI program with PGT-A analysis. Six eggs were fertilized out of 9 metaphase II oocytes. Five good quality blastocysts were submitted for genetic screening using 24-chromosome next generation sequencing (NGS). Two blastocysts were diagnosed as euploid and recommended for transfer. One euploid blastocyst was thawed and transferred to the patient’s uterus lining. Successful pregnancy was confirmed at 7 weeks of gestation with heartbeat. Successful delivery was achieved by Caesarean section at 38-39 weeks of gestation. Karyotyping demonstrated healthy genetic constitution of a baby. This case demonstrates a good evidence and potential of a transport scheme collaboration between IVF and genetic laboratories.
Collapse
|
21
|
Greco E, Litwicka K, Minasi MG, Cursio E, Greco PF, Barillari P. Preimplantation Genetic Testing: Where We Are Today. Int J Mol Sci 2020; 21:E4381. [PMID: 32575575 PMCID: PMC7352684 DOI: 10.3390/ijms21124381] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Revised: 06/13/2020] [Accepted: 06/16/2020] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Preimplantation genetic testing (PGT) is widely used today in in-vitro fertilization (IVF) centers over the world for selecting euploid embryos for transfer and to improve clinical outcomes in terms of embryo implantation, clinical pregnancy, and live birth rates. METHODS We report the current knowledge concerning these procedures and the results from different clinical indications in which PGT is commonly applied. RESULTS This paper illustrates different molecular techniques used for this purpose and the clinical significance of the different oocyte and embryo stage (polar bodies, cleavage embryo, and blastocyst) at which it is possible to perform sampling biopsies for PGT. Finally, genetic origin and clinical significance of embryo mosaicism are illustrated. CONCLUSIONS The preimplantation genetic testing is a valid technique to evaluated embryo euploidy and mosaicism before transfer.
Collapse
Affiliation(s)
- Ermanno Greco
- Reproductive Medicine, Villa Mafalda, 00199 Rome, Italy; (E.G.); (M.G.M.); (E.C.); (P.F.G.); (P.B.)
- UniCamillus, International Medical University, 00131 Rome, Italy
| | - Katarzyna Litwicka
- Reproductive Medicine, Villa Mafalda, 00199 Rome, Italy; (E.G.); (M.G.M.); (E.C.); (P.F.G.); (P.B.)
| | - Maria Giulia Minasi
- Reproductive Medicine, Villa Mafalda, 00199 Rome, Italy; (E.G.); (M.G.M.); (E.C.); (P.F.G.); (P.B.)
| | - Elisabetta Cursio
- Reproductive Medicine, Villa Mafalda, 00199 Rome, Italy; (E.G.); (M.G.M.); (E.C.); (P.F.G.); (P.B.)
| | - Pier Francesco Greco
- Reproductive Medicine, Villa Mafalda, 00199 Rome, Italy; (E.G.); (M.G.M.); (E.C.); (P.F.G.); (P.B.)
| | - Paolo Barillari
- Reproductive Medicine, Villa Mafalda, 00199 Rome, Italy; (E.G.); (M.G.M.); (E.C.); (P.F.G.); (P.B.)
| |
Collapse
|
22
|
Preimplantation Genetic Testing for Chromosomal Abnormalities: Aneuploidy, Mosaicism, and Structural Rearrangements. Genes (Basel) 2020; 11:genes11060602. [PMID: 32485954 PMCID: PMC7349251 DOI: 10.3390/genes11060602] [Citation(s) in RCA: 95] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Revised: 05/25/2020] [Accepted: 05/27/2020] [Indexed: 12/18/2022] Open
Abstract
There is a high incidence of chromosomal abnormalities in early human embryos, whether they are generated by natural conception or by assisted reproductive technologies (ART). Cells with chromosomal copy number deviations or chromosome structural rearrangements can compromise the viability of embryos; much of the naturally low human fecundity as well as low success rates of ART can be ascribed to these cytogenetic defects. Chromosomal anomalies are also responsible for a large proportion of miscarriages and congenital disorders. There is therefore tremendous value in methods that identify embryos containing chromosomal abnormalities before intrauterine transfer to a patient being treated for infertility—the goal being the exclusion of affected embryos in order to improve clinical outcomes. This is the rationale behind preimplantation genetic testing for aneuploidy (PGT-A) and structural rearrangements (-SR). Contemporary methods are capable of much more than detecting whole chromosome abnormalities (e.g., monosomy/trisomy). Technical enhancements and increased resolution and sensitivity permit the identification of chromosomal mosaicism (embryos containing a mix of normal and abnormal cells), as well as the detection of sub-chromosomal abnormalities such as segmental deletions and duplications. Earlier approaches to screening for chromosomal abnormalities yielded a binary result of normal versus abnormal, but the new refinements in the system call for new categories, each with specific clinical outcomes and nuances for clinical management. This review intends to give an overview of PGT-A and -SR, emphasizing recent advances and areas of active development.
Collapse
|
23
|
Macaulay AD, Allais A, FitzHarris G. Chromosome dynamics and spindle microtubule establishment in mouse embryos. FASEB J 2020; 34:8057-8067. [PMID: 32329130 DOI: 10.1096/fj.201902947r] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2019] [Revised: 03/25/2020] [Accepted: 03/31/2020] [Indexed: 11/11/2022]
Abstract
Chromosome segregation errors in mammalian embryos are common and jeopardize embryo health. Here, we perform for the first time 4-Dimensional imaging and tracking of chromosomes and centromeres through each preimplantation mitotic cell division in mouse embryos to define the normal dynamics of chromosome segregation. We show that a microtubule (MT)-dependent inward movement of chromosomes occurs at the time of nuclear envelope breakdown (NEBD), particularly in the earliest cell divisions, to position chromosomes prior to spindle assembly. Establishment of a rudimentary metaphase plate occurs immediately after NEBD, and is followed by a progressive alignment and biorientation of mitotic chromosomes. Stable end-on kinetochore-MT attachments form rapidly and attachment errors are uncommon. Altogether our data describe a rapid and efficient spindle assembly pathway that apparently minimizes the need for canonical MT attachment error correction in normally dividing embryos.
Collapse
Affiliation(s)
- Angus D Macaulay
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal, Montreal, QC, Canada
| | - Adélaïde Allais
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal, Montreal, QC, Canada
| | - Greg FitzHarris
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal, Montreal, QC, Canada.,Department of OBGYN, Université de Montréal, Montreal, QC, Canada
| |
Collapse
|
24
|
Popovic M, Dhaenens L, Boel A, Menten B, Heindryckx B. Chromosomal mosaicism in human blastocysts: the ultimate diagnostic dilemma. Hum Reprod Update 2020; 26:313-334. [DOI: 10.1093/humupd/dmz050] [Citation(s) in RCA: 62] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2019] [Revised: 11/29/2019] [Indexed: 12/30/2022] Open
Abstract
Abstract
BACKGROUND
Trophectoderm (TE) biopsy and next generation sequencing (NGS) are currently the preferred techniques for preimplantation genetic testing for aneuploidies (PGT-A). Although this approach delivered important improvements over previous testing strategies, increased sensitivity has also prompted a rise in diagnoses of uncertain clinical significance. This includes reports of chromosomal mosaicism, suggesting the presence of karyotypically distinct cells within a single TE biopsy. Given that PGT-A relies on the chromosomal constitution of the biopsied cells being representative of the entire embryo, the prevalence and clinical implications of blastocyst mosaicism continue to generate considerable controversy.
OBJECTIVE AND RATIONALE
The objective of this review was to evaluate existing scientific evidence regarding the prevalence and impact of chromosomal mosaicism in human blastocysts. We discuss insights from a biological, technical and clinical perspective to examine the implications of this diagnostic dilemma for PGT-A.
SEARCH METHODS
The PubMed and Google Scholar databases were used to search peer-reviewed publications using the following terms: ‘chromosomal mosaicism’, ‘human’, ‘embryo’, ‘blastocyst’, ‘implantation’, ‘next generation sequencing’ and ‘clinical management’ in combination with other keywords related to the subject area. Relevant articles in the English language, published until October 2019 were critically discussed.
OUTCOMES
Chromosomal mosaicism predominately results from errors in mitosis following fertilization. Although it appears to be less pervasive at later developmental stages, establishing the true prevalence of mosaicism in human blastocysts remains exceedingly challenging. In a clinical context, blastocyst mosaicism can only be reported based on a single TE biopsy and has been ascribed to 2–13% of embryos tested using NGS. Conversely, data from NGS studies disaggregating whole embryos suggests that mosaicism may be present in up to ~50% of blastocysts. However, differences in testing and reporting strategies, analysis platforms and the number of cells sampled inherently overshadow current data, while added uncertainties emanate from technical artefacts. Moreover, laboratory factors and aspects of in vitro culture generate further variability. Outcome data following the transfer of blastocysts diagnosed as mosaic remain limited. Current studies suggest that the transfer of putative mosaic embryos may lead to healthy live births, but also results in significantly reduced ongoing pregnancy rates compared to the transfer of euploid blastocysts. Observations that a subset of mosaic blastocysts has the capacity to develop normally have sparked discussions regarding the ability of embryos to self-correct. However, there is currently no direct evidence to support this assumption. Nevertheless, the exclusion of mosaic blastocysts results in fewer embryos available for transfer, which may inevitably compromise treatment outcomes.
WIDER IMPLICATIONS
Chromosomal mosaicism in human blastocysts remains a perpetual diagnostic and clinical dilemma in the context of PGT-A. This review offers an important scientific resource, informing about the challenges, risks and value of diagnosing mosaicism. Elucidating these uncertainties will ultimately pave the way towards improved clinical and patient management.
Collapse
Affiliation(s)
- Mina Popovic
- Ghent-Fertility and Stem Cell Team (G-FAST), Department for Reproductive Medicine, Ghent University Hospital, Corneel Heymanslaan 10, 9000 Ghent, Belgium
| | - Lien Dhaenens
- Ghent-Fertility and Stem Cell Team (G-FAST), Department for Reproductive Medicine, Ghent University Hospital, Corneel Heymanslaan 10, 9000 Ghent, Belgium
| | - Annekatrien Boel
- Ghent-Fertility and Stem Cell Team (G-FAST), Department for Reproductive Medicine, Ghent University Hospital, Corneel Heymanslaan 10, 9000 Ghent, Belgium
| | - Björn Menten
- Center for Medical Genetics, Ghent University Hospital, 9000 Ghent, Belgium
| | - Björn Heindryckx
- Ghent-Fertility and Stem Cell Team (G-FAST), Department for Reproductive Medicine, Ghent University Hospital, Corneel Heymanslaan 10, 9000 Ghent, Belgium
| |
Collapse
|
25
|
In vitro fertilization does not increase the incidence of de novo copy number alterations in fetal and placental lineages. Nat Med 2019; 25:1699-1705. [PMID: 31686035 DOI: 10.1038/s41591-019-0620-2] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2019] [Accepted: 09/20/2019] [Indexed: 01/21/2023]
Abstract
Although chromosomal instability (CIN) is a common phenomenon in cleavage-stage embryogenesis following in vitro fertilization (IVF)1-3, its rate in naturally conceived human embryos is unknown. CIN leads to mosaic embryos that contain a combination of genetically normal and abnormal cells, and is significantly higher in in vitro-produced preimplantation embryos as compared to in vivo-conceived preimplantation embryos4. Even though embryos with CIN-derived complex aneuploidies may arrest between the cleavage and blastocyst stages of embryogenesis5,6, a high number of embryos containing abnormal cells can pass this strong selection barrier7,8. However, neither the prevalence nor extent of CIN during prenatal development and at birth, following IVF treatment, is well understood. Here we profiled the genomic landscape of fetal and placental tissues postpartum from both IVF and naturally conceived children, to investigate the prevalence and persistence of large genetic aberrations that probably arose from IVF-related CIN. We demonstrate that CIN is not preserved at later stages of prenatal development, and that de novo numerical aberrations or large structural DNA imbalances occur at similar rates in IVF and naturally conceived live-born neonates. Our findings affirm that human IVF treatment has no detrimental effect on the chromosomal constitution of fetal and placental lineages.
Collapse
|
26
|
Munné S, Spinella F, Grifo J, Zhang J, Beltran MP, Fragouli E, Fiorentino F. Clinical outcomes after the transfer of blastocysts characterized as mosaic by high resolution Next Generation Sequencing- further insights. Eur J Med Genet 2019; 63:103741. [PMID: 31445143 DOI: 10.1016/j.ejmg.2019.103741] [Citation(s) in RCA: 68] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Revised: 05/28/2019] [Accepted: 08/13/2019] [Indexed: 12/01/2022]
Abstract
OBJECTIVE To determine the pregnancy outcome potential of euploid, mosaic and aneuploid embryos. DESIGN Retrospective study. SETTING Reference genetics laboratories. PATIENT(S) 2654 PGT-A cycles with euploid characterized embryo transfers, 253 PGT-A cycles with transfer of embryos characterized as mosaic, and 10 PGT-A cycles with fully abnormal embryo transfers. INTERVENTION(S) Blastocysts were assessed by trophectoderm (TE) biopsy followed by PGT-A via array CGH or NGS. MAIN OUTCOME MEASURE(S) Implantation, miscarriage, ongoing implantation rates (OIR), and karyotype if available, were compared between different embryo groups, and between the two PGT-A techniques. RESULTS The Ongoing Pregnancy Rate (OPR)/transfer was significantly higher for NGS-classified euploid embryos (85%) than for aCGH ones (71%) (p < 0.001), but the OPR/cycle was similar (63% vs 59%). NGS-classified mosaic embryos resulted in 37% OPR/cycle (p < 0.001 compared to euploid). Mosaic aneuploid embryos with <40% abnormal cells in the TE sample had an OIR of 50% compared to 27% for mosaics with 40-80% abnormal cells in the TE, and 9% for complex mosaic embryos. All the karyotyped ongoing pregnancies (n = 29) were euploid. Transfers of embryos classified as aneuploid via aCGH (n = 10) led to one chromosomally abnormal pregnancy. CONCLUSION(S) NGS-classified euploid embryos yielded higher OIRs but similar OPRs/cycle compared to aCGH. NGS-classified mosaic embryos had reduced potential to reach term, compared to euploid embryos. If they did reach term, those with karyotype results available were euploid. Embryos carrying uniform aneuploidies affecting entire chromosomes were mostly unable to implant after transfer, and the one that implanted ended up in a chromosomally abnormal live birth.
Collapse
Affiliation(s)
- Santiago Munné
- CooperGenomics, 3 Regent street, suite 301, Short Hills, NJ, USA; Overture Life, New York, NY, USA; Dept. OB/GYN, Yale University, New Haeven, CT, USA.
| | | | | | | | | | | | | |
Collapse
|
27
|
McCollin A, Swann RL, Summers MC, Handyside AH, Ottolini CS. Abnormal cleavage and developmental arrest of human preimplantation embryos in vitro. Eur J Med Genet 2019; 63:103651. [PMID: 30995534 DOI: 10.1016/j.ejmg.2019.04.008] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2019] [Revised: 04/01/2019] [Accepted: 04/09/2019] [Indexed: 02/08/2023]
Abstract
Despite improvements in culture conditions and laboratory techniques still only about 50% of human embryos reach the blastocyst stage of development in vitro. While many factors influence embryo development, aberrant cleavage divisions have only recently been shown to directly affect the genome in individual cells of human embryos resulting in chromosome loss, mosaicism and cell arrest. In this article we review the current literature in the area of aberrant cleavage in human embryos and its effect on blastocyst development. Further to this, we propose a series of common abnormal cleavage events, with particular attention to timing and frequency, and illustrate how these might influence a number of different embryo fates.
Collapse
Affiliation(s)
- Abeo McCollin
- London Women's Clinic, One St Thomas Street, London, SE1 9RY, UK
| | | | - Michael C Summers
- London Women's Clinic, One St Thomas Street, London, SE1 9RY, UK; School of Biosciences, University of Kent, Canterbury, CT2 7NJ, UK
| | - Alan H Handyside
- London Women's Clinic, One St Thomas Street, London, SE1 9RY, UK; School of Biosciences, University of Kent, Canterbury, CT2 7NJ, UK
| | - Christian S Ottolini
- The Evewell, 61 Harley Street, London, W1G 8QU, UK; School of Biosciences, University of Kent, Canterbury, CT2 7NJ, UK.
| |
Collapse
|
28
|
Fowler KE, Mandawala AA, Griffin DK. The role of chromosome segregation and nuclear organisation in human subfertility. Biochem Soc Trans 2019; 47:425-432. [DOI: 10.1042/bst20180231] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/30/2023]
Abstract
Abstract
Spermatogenesis is central to successful sexual reproduction, producing large numbers of haploid motile male gametes. Throughout this process, a series of equational and reductional chromosome segregation precedes radical repackaging of the haploid genome. Faithful chromosome segregation is thus crucial, as is an ordered spatio-temporal ‘dance’ of packing a large amount of chromatin into a very small space. Ergo, when the process goes wrong, this is associated with an improper chromosome number, nuclear position and/or chromatin damage in the sperm head. Generally, screening for overall DNA damage is relatively commonplace in clinics, but aneuploidy assessment is less so and nuclear organisation studies form the basis of academic research. Several studies have focussed on the role of chromosome segregation, nuclear organisation and analysis of sperm morphometry in human subfertility observing significant alterations in some cases, especially of the sex chromosomes. Importantly, sperm DNA damage has been associated with infertility and both extrinsic (e.g. lifestyle) and intrinsic (e.g. reactive oxygen species levels) factors, and while some DNA-strand breaks are repaired, unexpected breaks can cause differential chromatin packaging and further breakage. A ‘healthy’ sperm nucleus (with the right number of chromosomes, nuclear organisation and minimal DNA damage) is thus an essential part of reproduction. The purpose of this review is to summarise state of the art in the fields of sperm aneuploidy assessment, nuclear organisation and DNA damage studies.
Collapse
Affiliation(s)
- Katie E. Fowler
- School of Human and Life Sciences, Canterbury Christ Church University, Canterbury, U.K
| | - Anjali A. Mandawala
- School of Human and Life Sciences, Canterbury Christ Church University, Canterbury, U.K
| | | |
Collapse
|
29
|
Alteri A, Corti L, Sanchez AM, Rabellotti E, Papaleo E, Viganò P. Assessment of pre-implantation genetic testing for embryo aneuploidies: A SWOT analysis. Clin Genet 2019; 95:479-487. [DOI: 10.1111/cge.13510] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2018] [Revised: 12/29/2018] [Accepted: 01/04/2019] [Indexed: 12/25/2022]
Affiliation(s)
- Alessandra Alteri
- Obstetrics and Gynaecology Department; IRCCS San Raffaele Scientific Institute; Milan Italy
| | - Laura Corti
- Obstetrics and Gynaecology Department; IRCCS San Raffaele Scientific Institute; Milan Italy
| | - Ana M. Sanchez
- Reproductive Sciences Laboratory; Division of Genetics and Cell Biology, IRCCS San Raffaele Scientific Institute; Milan Italy
| | - Elisa Rabellotti
- Obstetrics and Gynaecology Department; IRCCS San Raffaele Scientific Institute; Milan Italy
| | - Enrico Papaleo
- Obstetrics and Gynaecology Department; IRCCS San Raffaele Scientific Institute; Milan Italy
- Reproductive Sciences Laboratory; Division of Genetics and Cell Biology, IRCCS San Raffaele Scientific Institute; Milan Italy
| | - Paola Viganò
- Reproductive Sciences Laboratory; Division of Genetics and Cell Biology, IRCCS San Raffaele Scientific Institute; Milan Italy
| |
Collapse
|
30
|
Chunduri NK, Storchová Z. The diverse consequences of aneuploidy. Nat Cell Biol 2019; 21:54-62. [PMID: 30602769 DOI: 10.1038/s41556-018-0243-8] [Citation(s) in RCA: 132] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2018] [Accepted: 10/31/2018] [Indexed: 12/25/2022]
Abstract
Aneuploidy, or imbalanced chromosome number, has profound effects on eukaryotic cells. In humans, aneuploidy is associated with various pathologies, including cancer, which suggests that it mediates a proliferative advantage under these conditions. Here, we discuss physiological changes triggered by aneuploidy, such as altered cell growth, transcriptional changes, proteotoxic stress, genomic instability and response to interferons, and how cancer cells adapt to the changing aneuploid genome.
Collapse
Affiliation(s)
| | - Zuzana Storchová
- Department of Molecular Genetics, TU Kaiserslautern, Kaiserslautern, Germany.
| |
Collapse
|
31
|
Nakhuda G, Jing C, Butler R, Guimond C, Hitkari J, Taylor E, Tallon N, Yuzpe A. Frequencies of chromosome-specific mosaicisms in trophoectoderm biopsies detected by next-generation sequencing. Fertil Steril 2018; 109:857-865. [PMID: 29778385 DOI: 10.1016/j.fertnstert.2018.01.011] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2017] [Revised: 01/02/2018] [Accepted: 01/08/2018] [Indexed: 11/30/2022]
Abstract
OBJECTIVE To examine the chromosome-specific frequencies of mosaicism detected by next-generation sequencing (NGS) compared with constitutional aneuploidy. DESIGN Retrospective cross-sectional review of NGS results from trophectoderm biopsies analyzed by per-chromosome prevalence of mosaicism and constitutional aneuploidy. SETTING Private fertility clinic. PATIENT(S) A total of 378 patients who underwent preimplantation genetic screening by NGS for routine clinical indications from February 2016 to April 2017. INTERVENTION(S) None. MAIN OUTCOME MEASURE(S) Aneuploidies and mosaicisms were tabulated per chromosome, and whole-chromosome and segmental mosaicisms were also analyzed. RESULT(S) NGS results were analyzed from 1,547 blastocysts. Mosaicism was detected as the sole abnormality in 17.5% (n = 270) of samples but were also found in 196/634 aneuploid embryos, so the overall incidence of mosaicism per biopsy was 30.1%. Mosaicism did not statistically vary when stratified by maternal age. The mean rate of overall mosaicism per chromosome was 2.46%. When whole chromosome and segmental mosaicisms were compared, unequal frequencies were found in several chromosomes. Trisomy was more frequently detected as whole-chromosome mosaicism, although monosomy was more frequently seen in segmental mosaicism. Aneuploidy and mosaicism displayed different patterns of distribution in various chromosomes. CONCLUSION(S) Mosaicism is unequally detected in various chromosomes and appears distinct from the distribution pattern of constitutional aneuploidy. Whole chromosome and segmental mosaicisms are also differentially detected. These results contribute to the study of mosaicism, illuminating a differential pattern of detection across the genome.
Collapse
Affiliation(s)
- Gary Nakhuda
- Olive Fertility Centre, Vancouver, British Columbia, Canada; University of British Columbia, Vancouver, British Columbia, Canada.
| | - Chen Jing
- Olive Fertility Centre, Vancouver, British Columbia, Canada
| | - Rachel Butler
- Olive Fertility Centre, Vancouver, British Columbia, Canada; University of British Columbia, Vancouver, British Columbia, Canada
| | - Colleen Guimond
- Olive Fertility Centre, Vancouver, British Columbia, Canada; University of British Columbia, Vancouver, British Columbia, Canada
| | - Jason Hitkari
- Olive Fertility Centre, Vancouver, British Columbia, Canada; University of British Columbia, Vancouver, British Columbia, Canada
| | - Elizabeth Taylor
- Olive Fertility Centre, Vancouver, British Columbia, Canada; University of British Columbia, Vancouver, British Columbia, Canada
| | - Niamh Tallon
- Olive Fertility Centre, Vancouver, British Columbia, Canada; University of British Columbia, Vancouver, British Columbia, Canada
| | - Albert Yuzpe
- Olive Fertility Centre, Vancouver, British Columbia, Canada; University of British Columbia, Vancouver, British Columbia, Canada
| |
Collapse
|
32
|
Griffin DK, Ogur C. Chromosomal analysis in IVF: just how useful is it? Reproduction 2018; 156:F29-F50. [PMID: 29945889 DOI: 10.1530/rep-17-0683] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2017] [Accepted: 03/23/2018] [Indexed: 12/11/2022]
Abstract
Designed to minimize chances of genetically abnormal embryos, preimplantation genetic diagnosis (PGD) involves in vitro fertilization (IVF), embryo biopsy, diagnosis and selective embryo transfer. Preimplantation genetic testing for aneuploidy (PGT-A) aims to avoid miscarriage and live born trisomic offspring and to improve IVF success. Diagnostic approaches include fluorescence in situ hybridization (FISH) and more contemporary comprehensive chromosome screening (CCS) including array comparative genomic hybridization (aCGH), quantitative polymerase chain reaction (PCR), next-generation sequencing (NGS) and karyomapping. NGS has an improved dynamic range, and karyomapping can detect chromosomal and monogenic disorders simultaneously. Mosaicism (commonplace in human embryos) can arise by several mechanisms; those arising initially meiotically (but with a subsequent post-zygotic 'trisomy rescue' event) usually lead to adverse outcomes, whereas the extent to which mosaics that are initially chromosomally normal (but then arise purely post-zygotically) can lead to unaffected live births is uncertain. Polar body (PB) biopsy is the least common sampling method, having drawbacks including cost and inability to detect any paternal contribution. Historically, cleavage-stage (blastomere) biopsy has been the most popular; however, higher abnormality levels, mosaicism and potential for embryo damage have led to it being superseded by blastocyst (trophectoderm - TE) biopsy, which provides more cells for analysis. Improved biopsy, diagnosis and freeze-all strategies collectively have the potential to revolutionize PGT-A, and there is increasing evidence of their combined efficacy. Nonetheless, PGT-A continues to attract criticism, prompting questions of when we consider the evidence base sufficient to justify routine PGT-A? Basic biological research is essential to address unanswered questions concerning the chromosome complement of human embryos, and we thus entreat companies, governments and charities to fund more. This will benefit both IVF patients and prospective parents at risk of aneuploid offspring following natural conception. The aim of this review is to appraise the 'state of the art' in terms of PGT-A, including the controversial areas, and to suggest a practical 'way forward' in terms of future diagnosis and applied research.
Collapse
Affiliation(s)
- Darren K Griffin
- School of BiosciencesCentre for Interdisciplinary Studies of Reproduction, University of Kent, Canterbury, UK
| | - Cagri Ogur
- Bahceci Genetic Diagnosis Centerİstanbul, Turkey.,Department of BioengineeringYildiz Technical University, İstanbul, Turkey
| |
Collapse
|
33
|
Should every embryo undergo preimplantation genetic testing for aneuploidy? A review of the modern approach to in vitro fertilization. Best Pract Res Clin Obstet Gynaecol 2018; 53:38-47. [DOI: 10.1016/j.bpobgyn.2018.07.005] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2018] [Accepted: 07/17/2018] [Indexed: 11/19/2022]
|
34
|
Zhang L, Wei D, Zhu Y, Gao Y, Yan J, Chen ZJ. Rates of live birth after mosaic embryo transfer compared with euploid embryo transfer. J Assist Reprod Genet 2018; 36:165-172. [PMID: 30246223 DOI: 10.1007/s10815-018-1322-2] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2018] [Accepted: 09/14/2018] [Indexed: 12/22/2022] Open
Abstract
PURPOSE Mosaicism is a prevalent characteristic of human preimplantation embryos. This retrospective cohort study aimed to investigate pregnancy outcomes after transfer of mosaic or euploid embryos. METHODS The embryos, which had been transferred as "euploidy," were processed using array-based comparative genomic hybridization (aCGH). The original aCGH charts of the transferred embryos were reanalyzed. Mosaic and control euploid embryos were defined according to log2 ratio calls. RESULTS Overall, 102 embryos were determined to be mosaic, of which 101 were estimated to harbor no more than 50% aneuploid mosaicism. Additionally, 268 euploid embryos were matched as controls. The rates of live birth (46.6% vs. 59.1%, odds ratio (OR) 0.60, 95% confidence interval (CI) 0.38-0.95), and biochemical pregnancy (65.7% vs. 76.1%, OR 0.60, 95% CI 0.37-0.99) per transfer cycle were significantly lower after mosaic embryo transfer than after euploid embryo transfer. The rates of clinical pregnancy and pregnancy loss and the risks of obstetric outcomes did not differ significantly between the two groups. CONCLUSIONS Compared with euploid embryo transfer, mosaic embryo transfer is associated with a lower rate of live birth, which is mainly attributed to a decreased rate of conception. However, as mosaic embryo transfer yielded a live birth rate of 46.6%, patients without euploid embryos could be counseled regarding this alternative option.
Collapse
Affiliation(s)
- Lei Zhang
- Center for Reproductive Medicine, Shandong Provincial Hospital Affiliated to Shandong University, Jingliu Road 157, Jinan, 250021, China.,National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Jinan, China.,The Key Laboratory of Reproductive Endocrinology (Shandong University), Ministry of Education, Jinan, China.,Shandong Provincial Key Laboratory of Reproductive Medicine, Jinan, China
| | - Daimin Wei
- Center for Reproductive Medicine, Shandong Provincial Hospital Affiliated to Shandong University, Jingliu Road 157, Jinan, 250021, China.,National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Jinan, China.,The Key Laboratory of Reproductive Endocrinology (Shandong University), Ministry of Education, Jinan, China.,Shandong Provincial Key Laboratory of Reproductive Medicine, Jinan, China
| | - Yueting Zhu
- Center for Reproductive Medicine, Shandong Provincial Hospital Affiliated to Shandong University, Jingliu Road 157, Jinan, 250021, China.,National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Jinan, China.,The Key Laboratory of Reproductive Endocrinology (Shandong University), Ministry of Education, Jinan, China.,Shandong Provincial Key Laboratory of Reproductive Medicine, Jinan, China
| | - Yuan Gao
- Center for Reproductive Medicine, Shandong Provincial Hospital Affiliated to Shandong University, Jingliu Road 157, Jinan, 250021, China.,National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Jinan, China.,The Key Laboratory of Reproductive Endocrinology (Shandong University), Ministry of Education, Jinan, China.,Shandong Provincial Key Laboratory of Reproductive Medicine, Jinan, China
| | - Junhao Yan
- Center for Reproductive Medicine, Shandong Provincial Hospital Affiliated to Shandong University, Jingliu Road 157, Jinan, 250021, China. .,National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Jinan, China. .,The Key Laboratory of Reproductive Endocrinology (Shandong University), Ministry of Education, Jinan, China. .,Shandong Provincial Key Laboratory of Reproductive Medicine, Jinan, China.
| | - Zi-Jiang Chen
- Center for Reproductive Medicine, Shandong Provincial Hospital Affiliated to Shandong University, Jingliu Road 157, Jinan, 250021, China.,National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Jinan, China.,The Key Laboratory of Reproductive Endocrinology (Shandong University), Ministry of Education, Jinan, China.,Shandong Provincial Key Laboratory of Reproductive Medicine, Jinan, China.,Center for Reproductive Medicine, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.,Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai, China
| |
Collapse
|
35
|
Abstract
DNA mutations as a consequence of errors during DNA damage repair, replication, or mitosis are the substrate for evolution. In multicellular organisms, mutations can occur in the germline and also in somatic tissues, where they are associated with cancer and other chronic diseases and possibly with aging. Recent advances in high-throughput sequencing have made it relatively easy to study germline de novo mutations, but in somatic cells, the vast majority of mutations are low-abundant and can be detected only in clonal lineages, such as tumors, or single cells. Here we review recent results on somatic mutations in normal human and animal tissues with a focus on their possible functional consequences.
Collapse
Affiliation(s)
- Lei Zhang
- Department of Genetics, Albert Einstein College of Medicine, Bronx, New York 10461, USA;
| | - Jan Vijg
- Department of Genetics, Albert Einstein College of Medicine, Bronx, New York 10461, USA;
| |
Collapse
|
36
|
Vázquez-Diez C, FitzHarris G. Causes and consequences of chromosome segregation error in preimplantation embryos. Reproduction 2018; 155:R63-R76. [DOI: 10.1530/rep-17-0569] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2017] [Revised: 10/19/2017] [Accepted: 11/06/2017] [Indexed: 01/04/2023]
Abstract
Errors in chromosome segregation are common during the mitotic divisions of preimplantation development in mammalian embryos, giving rise to so-called ‘mosaic’ embryos possessing a mixture of euploid and aneuploid cells. Mosaicism is widely considered to be detrimental to embryo quality and is frequently used as criteria to select embryos for transfer in human fertility clinics. However, despite the clear clinical importance, the underlying defects in cell division that result in mosaic aneuploidy remain elusive. In this review, we summarise recent findings from clinical and animal model studies that provide new insights into the fundamental mechanisms of chromosome segregation in the highly unusual cellular environment of early preimplantation development and consider recent clues as to why errors should commonly occur in this setting. We furthermore discuss recent evidence suggesting that mosaicism is not an irrevocable barrier to a healthy pregnancy. Understanding the causes and biological impacts of mosaic aneuploidy will be pivotal in the development and fine-tuning of clinical embryo selection methods.
Collapse
|
37
|
Abstract
Preimplantation genetic diagnosis was first successfully performed in 1989 as an alternative to prenatal diagnosis for couples at risk of transmitting a genetic or chromosomal abnormality, such as cystic fibrosis, to their child. From embryos generated in vitro, biopsied cells are genetically tested. From the mid-1990s, this technology has been employed as an embryo selection tool for patients undergoing in vitro fertilisation, screening as many chromosomes as possible, in the hope that selecting chromosomally normal embryos will lead to higher implantation and decreased miscarriage rates. This procedure, preimplantation genetic screening, was initially performed using fluorescent in situ hybridisation, but 11 randomised controlled trials of screening using this technique showed no improvement in in vitro fertilisation delivery rates. Progress in genetic testing has led to the introduction of array comparative genomic hybridisation, quantitative polymerase chain reaction, and next generation sequencing for preimplantation genetic screening, and three small randomised controlled trials of preimplantation genetic screening using these new techniques indicate a modest benefit. Other trials are still in progress but, regardless of their results, preimplantation genetic screening is now being offered globally. In the near future, it is likely that sequencing will be used to screen the full genetic code of the embryo.
Collapse
Affiliation(s)
- Joyce C Harper
- Joyce Harper, Embryology, IVF and Reproductive Genetics Group, Institute for Women's Health, University College London, London, UK
| |
Collapse
|
38
|
Munné S, Blazek J, Large M, Martinez-Ortiz PA, Nisson H, Liu E, Tarozzi N, Borini A, Becker A, Zhang J, Maxwell S, Grifo J, Babariya D, Wells D, Fragouli E. Detailed investigation into the cytogenetic constitution and pregnancy outcome of replacing mosaic blastocysts detected with the use of high-resolution next-generation sequencing. Fertil Steril 2017; 108:62-71.e8. [PMID: 28579407 DOI: 10.1016/j.fertnstert.2017.05.002] [Citation(s) in RCA: 182] [Impact Index Per Article: 22.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2017] [Revised: 04/26/2017] [Accepted: 05/03/2017] [Indexed: 11/25/2022]
Abstract
OBJECTIVE To determine the pregnancy outcome potential of mosaic embryos, detected by means of preimplantation genetic screening (PGS) with the use of next-generation sequencing (NGS). DESIGN Retrospective study. SETTING Genetics laboratories. PATIENT(S) PGS cycles during which either mosaic or euploid embryos were replaced. INTERVENTION(S) Blastocysts were biopsied and processed with the use of NGS, followed by frozen embryo transfer. Trophectoderm (TE) biopsies were classified as mosaic if they had 20%-80% abnormal cells. MAIN OUTCOME MEASURE(S) Implantation, miscarriage rates, and ongoing implantation rates (OIRs) were compared between euploid and types of mosaic blastocysts. RESULT(S) Complex mosaic embryos had a significantly lower OIR (10%) than aneuploidy mosaic (50%), double aneuploidy mosaic (45%), and segmental mosaic (41%). There was a tendency for mosaics with 40%-80% abnormal cells to have a lower OIR than those with <40% (22% vs. 56%). However, few embryos (n = 34) with a mosaic error in 40%-80% of the TE sample were replaced. There was no difference between monosomic and trisomic mosaics or between entire chromosome mosaicism or segmental mosaicism. Implantation rates were significantly higher (70% vs. 53%), miscarriage rates lower (10% vs. 25%), and OIRs higher (63% vs. 40%) after euploid embryo transfer than after mosaic embryo transfer. CONCLUSION(S) Forty-one percent of mosaic embryos produced an ongoing implantation. Complex mosaic blastocysts had a lower OIR than other mosaics. Mosaic monosomies performed as well as mosaic trisomies and mosaic segmental aneuploidies. The results suggest that embryos with >40% abnormal cells and those with multiple mosaic abnormalities (chaotic mosaics) are likely to have lower OIRs and should be given low transfer priority.
Collapse
Affiliation(s)
- Santiago Munné
- Reprogenetics (Cooper Genomics), Livingston, New Jersey.
| | | | | | | | - Haley Nisson
- Reprogenetics (Cooper Genomics), Livingston, New Jersey
| | - Emmeline Liu
- Reprogenetics (Cooper Genomics), Livingston, New Jersey
| | | | | | | | | | | | | | | | - Dagan Wells
- Reprogenetics (Cooper Genomics), Oxford, United Kingdom
| | | |
Collapse
|
39
|
Abstract
INTRODUCTION Preimplantation genetic diagnosis and screening (PGD/PGS) has been applied clinically for >25 years however inherent drawbacks include the necessity to tailor each case to the trait in question, and that technology to detect monogenic and chromosomal disorders respectively is fundamentally different. Areas covered: The area of preimplantation genetics has evolved over the last 25 years, adapting to changes in technology and the need for more efficient, streamlined diagnoses. Karyomapping allows the determination of inheritance from the (grand)parental haplobocks through assembly of inherited chromosomal segments. The output displays homologous chromosomes, crossovers and the genetic status of the embryos by linkage comparison, as well as chromosomal disorders. It also allows for determination of heterozygous SNP calls, avoiding the risks of allele dropout, a common problem with other PGD techniques. Manuscripts documenting the evolution of preimplantation genetics, especially those investigating technologies that would simultaneously detect monogenic and chromosomal disorders, were selected for review. Expert commentary: Karyomapping is currently available for detection of single gene disorders; ~1000 clinics worldwide offer it (via ~20 diagnostic laboratories) and ~2500 cases have been performed. Due an inability to detect post-zygotic trisomy reliably however and confounding problems of embryo mosaicism, karyomapping has yet to be applied clinically for detection of chromosome disorders.
Collapse
Affiliation(s)
- Rebecca L Gould
- a The Bridge Centre , London , UK.,b School of Biological Sciences , University of Kent , Canterbury , UK
| | - Darren K Griffin
- b School of Biological Sciences , University of Kent , Canterbury , UK
| |
Collapse
|
40
|
Munné S, Wells D. Detection of mosaicism at blastocyst stage with the use of high-resolution next-generation sequencing. Fertil Steril 2017; 107:1085-1091. [DOI: 10.1016/j.fertnstert.2017.03.024] [Citation(s) in RCA: 94] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2017] [Accepted: 03/24/2017] [Indexed: 01/07/2023]
|
41
|
McCoy RC. Mosaicism in Preimplantation Human Embryos: When Chromosomal Abnormalities Are the Norm. Trends Genet 2017; 33:448-463. [PMID: 28457629 DOI: 10.1016/j.tig.2017.04.001] [Citation(s) in RCA: 154] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2016] [Revised: 03/31/2017] [Accepted: 04/03/2017] [Indexed: 11/15/2022]
Abstract
Along with errors in meiosis, mitotic errors during post-zygotic cell division contribute to pervasive aneuploidy in human embryos. Relatively little is known, however, about the genesis of these errors or their fitness consequences. Rapid technological advances are helping to close this gap, revealing diverse molecular mechanisms contributing to mitotic error. These include altered cell cycle checkpoints, aberrations of the centrosome, and failed chromatid cohesion, mirroring findings from cancer biology. Recent studies are challenging the idea that mitotic error is abnormal, emphasizing that the fitness impacts of mosaicism depend on its scope and severity. In light of these findings, technical and philosophical limitations of various screening approaches are discussed, along with avenues for future research.
Collapse
Affiliation(s)
- Rajiv C McCoy
- Department of Genome Sciences, University of Washington, Seattle, WA, USA.
| |
Collapse
|
42
|
Sanders KD, Griffin DK. Chromosomal Preimplantation Genetic Diagnosis: 25 Years and Counting. JOURNAL OF FETAL MEDICINE 2017. [DOI: 10.1007/s40556-017-0123-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
|
43
|
Fragouli E, Alfarawati S, Spath K, Babariya D, Tarozzi N, Borini A, Wells D. Analysis of implantation and ongoing pregnancy rates following the transfer of mosaic diploid–aneuploid blastocysts. Hum Genet 2017; 136:805-819. [DOI: 10.1007/s00439-017-1797-4] [Citation(s) in RCA: 148] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2017] [Accepted: 03/09/2017] [Indexed: 11/24/2022]
|
44
|
Sachdev NM, Maxwell SM, Besser AG, Grifo JA. Diagnosis and clinical management of embryonic mosaicism. Fertil Steril 2017; 107:6-11. [DOI: 10.1016/j.fertnstert.2016.10.006] [Citation(s) in RCA: 59] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2016] [Revised: 09/30/2016] [Accepted: 10/03/2016] [Indexed: 11/16/2022]
|
45
|
Braakhekke M, Mol F, Mastenbroek S, Mol BWJ, van der Veen F. Equipoise and the RCT. Hum Reprod 2016; 32:257-260. [DOI: 10.1093/humrep/dew286] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2016] [Revised: 10/13/2016] [Accepted: 10/26/2016] [Indexed: 11/13/2022] Open
|
46
|
Kane SC, Willats E, Bezerra Maia e Holanda Moura S, Hyett J, da Silva Costa F. Pre-Implantation Genetic Screening Techniques: Implications for Clinical Prenatal Diagnosis. Fetal Diagn Ther 2016; 40:241-254. [DOI: 10.1159/000449381] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2016] [Accepted: 08/23/2016] [Indexed: 11/19/2022]
|
47
|
Shamonki MI, Jin H, Haimowitz Z, Liu L. Proof of concept: preimplantation genetic screening without embryo biopsy through analysis of cell-free DNA in spent embryo culture media. Fertil Steril 2016; 106:1312-1318. [PMID: 27565258 DOI: 10.1016/j.fertnstert.2016.07.1112] [Citation(s) in RCA: 94] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2016] [Revised: 07/05/2016] [Accepted: 07/21/2016] [Indexed: 11/29/2022]
Abstract
OBJECTIVE To assess whether preimplantation genetic screening (PGS) is possible by testing for free embryonic DNA in spent IVF media from embryos undergoing trophectoderm biopsy. DESIGN Prospective cohort analysis. SETTING Academic fertility center. PATIENT(S) Seven patients undergoing IVF and 57 embryos undergoing trophectoderm biopsy for PGS. INTERVENTION(S) On day 3 of development, each embryo was placed in a separate media droplet. All biopsied embryos received a PGS result by array comparative genomic hybridization. Preimplantation genetic screening was performed on amplified DNA extracted from media and results were compared with PGS results for the corresponding biopsy. MAIN OUTCOME MEASURE(S) [1] Presence of DNA in spent IVF culture media. [2] Correlation between genetic screening result from spent media and corresponding biopsy. RESULT(S) Fifty-five samples had detectable DNA ranging from 2-642 ng/μL after a 2-hour amplification. Six samples with the highest DNA levels underwent PGS, rendering one result with a derivative log ratio SD (DLRSD) of <0.85 (a quality control metric of oligonucleotide array comparative genomic hybridization). The fluid sample and trophectoderm results were identical demonstrating (45XY, -13). Three samples were reamplified 1 hour later and tested showing improving DLRSD. One of the three samples with a DLRSD of 0.85 demonstrated (46XY), consistent with the biopsy. Overnight DNA amplification showed DNA in all samples. CONCLUSION(S) We demonstrate two novel findings: the presence of free embryonic DNA in spent media and a result that is consistent with trophectoderm biopsy. Improvements in DNA collection, amplification, and testing may allow for PGS without biopsy in the future.
Collapse
Affiliation(s)
- Mousa I Shamonki
- Fertility and Surgical Associates of California, Thousand Oaks, California; University of California, Los Angeles, Fertility and Reproductive Health Center, Los Angeles, California.
| | - Helen Jin
- PacGenomics, Agoura Hills, California
| | | | - Lian Liu
- PacGenomics, Agoura Hills, California
| |
Collapse
|
48
|
Denomme MM, McCallie BR, Parks JC, Schoolcraft WB, Katz-Jaffe MG. Epigenetic Dysregulation Observed in Monosomy Blastocysts Further Compromises Developmental Potential. PLoS One 2016; 11:e0156980. [PMID: 27271036 PMCID: PMC4896457 DOI: 10.1371/journal.pone.0156980] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2016] [Accepted: 05/23/2016] [Indexed: 01/23/2023] Open
Abstract
Epigenetic mechanisms such as DNA methylation regulate genomic imprinting and account for the distinct non-equivalence of the parental genomes in the embryo. Chromosomal aneuploidy, a major cause of infertility, distorts this highly regulated disparity by the presence or absence of chromosomes. The implantation potential of monosomy embryos is negligible compared to their trisomy counterparts, yet the cause for this is unknown. This study investigated the impact of chromosomal aneuploidy on strict epigenetically regulated domains, specifically imprinting control regions present on aneuploid chromosomes. Donated cryopreserved human IVF blastocysts of transferable quality, including trisomy 15, trisomy 11, monosomy 15, monosomy 11, and donor oocyte control blastocysts were examined individually for DNA methylation profiles by bisulfite mutagenesis and sequencing analysis of two maternally methylated imprinting control regions (ICRs), SNRPN (15q11.2) and KCNQ1OT1 (11p15.5), and one paternally methylated imprinting control region, H19 (11p15.5). Imprinted genes within the regions were also evaluated for transcript abundance by RT-qPCR. Overall, statistically significant hypermethylated and hypomethylated ICRs were found in both the trisomy and monosomy blastocysts compared to controls, restricted only to the chromosome affected by the aneuploidy. Increased expression was observed for maternally-expressed imprinted genes in trisomy blastocysts, while a decreased expression was observed for both maternally- and paternally-expressed imprinted genes in monosomy blastocysts. This epigenetic dysregulation and altered monoallelic expression observed at imprinting control regions in aneuploid IVF embryos supports euploid embryo transfer during infertility treatments, and may specifically highlight an explanation for the compromised implantation potential in monosomy embryos.
Collapse
Affiliation(s)
| | - Blair R. McCallie
- Fertility Labs of Colorado, Lone Tree, Colorado, United States of America
| | - Jason C. Parks
- Fertility Labs of Colorado, Lone Tree, Colorado, United States of America
| | - William B. Schoolcraft
- Colorado Center for Reproductive Medicine, Lone Tree, Colorado, United States of America
| | - Mandy G. Katz-Jaffe
- Fertility Labs of Colorado, Lone Tree, Colorado, United States of America
- Colorado Center for Reproductive Medicine, Lone Tree, Colorado, United States of America
- * E-mail:
| |
Collapse
|
49
|
Daughtry BL, Chavez SL. Chromosomal instability in mammalian pre-implantation embryos: potential causes, detection methods, and clinical consequences. Cell Tissue Res 2016; 363:201-225. [PMID: 26590822 PMCID: PMC5621482 DOI: 10.1007/s00441-015-2305-6] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2015] [Accepted: 09/24/2015] [Indexed: 01/08/2023]
Abstract
Formation of a totipotent blastocyst capable of implantation is one of the first major milestones in early mammalian embryogenesis, but less than half of in vitro fertilized embryos from most mammals will progress to this stage of development. Whole chromosomal abnormalities, or aneuploidy, are key determinants of whether human embryos will arrest or reach the blastocyst stage. Depending on the type of chromosomal abnormality, however, certain embryos still form blastocysts and may be morphologically indistinguishable from chromosomally normal embryos. Despite the implementation of pre-implantation genetic screening and other advanced in vitro fertilization (IVF) techniques, the identification of aneuploid embryos remains complicated by high rates of mosaicism, atypical cell division, cellular fragmentation, sub-chromosomal instability, and micro-/multi-nucleation. Moreover, several of these processes occur in vivo following natural human conception, suggesting that they are not simply a consequence of culture conditions. Recent technological achievements in genetic, epigenetic, chromosomal, and non-invasive imaging have provided additional embryo assessment approaches, particularly at the single-cell level, and clinical trials investigating their efficacy are continuing to emerge. In this review, we summarize the potential mechanisms by which aneuploidy may arise, the various detection methods, and the technical advances (such as time-lapse imaging, "-omic" profiling, and next-generation sequencing) that have assisted in obtaining this data. We also discuss the possibility of aneuploidy resolution in embryos via various corrective mechanisms, including multi-polar divisions, fragment resorption, endoreduplication, and blastomere exclusion, and conclude by examining the potential implications of these findings for IVF success and human fecundity.
Collapse
Affiliation(s)
- Brittany L Daughtry
- Department of Cell, Developmental & Cancer Biology, Graduate Program in Molecular & Cellular Biosciences, Oregon Health & Science University School of Medicine, Portland, Ore., USA
- Division of Reproductive & Developmental Sciences, Oregon National Primate Research Center, 505 NW 185th Avenue, Beaverton, OR 97006, USA
| | - Shawn L Chavez
- Division of Reproductive & Developmental Sciences, Oregon National Primate Research Center, 505 NW 185th Avenue, Beaverton, OR 97006, USA.
- Physiology & Pharmacology, Oregon Health & Science University School of Medicine, Portland, Ore., USA.
- Department of Obstetrics & Gynecology, Oregon Health & Science University School of Medicine, Portland, Ore., USA.
| |
Collapse
|
50
|
Luna D, Hilario R, Dueñas-Chacón J, Romero R, Zavala P, Villegas L, García-Ferreyra J. The IMSI Procedure Improves Laboratory and Clinical Outcomes Without Compromising the Aneuploidy Rate When Compared to the Classical ICSI Procedure. CLINICAL MEDICINE INSIGHTS. REPRODUCTIVE HEALTH 2015; 9:29-37. [PMID: 26609251 PMCID: PMC4644143 DOI: 10.4137/cmrh.s33032] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/10/2015] [Revised: 10/13/2015] [Accepted: 10/14/2015] [Indexed: 12/20/2022]
Abstract
PURPOSE The intracytoplasmic morphologically selected sperm injection (IMSI) procedure has been associated with better laboratory and clinical outcomes in assisted reproduction technologies. Less information is available regarding the relationship between embryo aneuploidy rate and the IMSI procedure. The aim of this study is to compare the clinical outcomes and chromosomal status of IMSI-derived embryos with those obtained from intracytoplasmic sperm injection (ICSI) in order to establish a clearer view of the benefits of IMSI in infertile patients. METHODS We retrospectively analyzed a total of 11 cycles of IMSI and 20 cycles of ICSI with preimplantation genetic diagnosis. The fertilization rate, cleavage rate, embryo quality, blastocyst development, aneuploidy rate, pregnancy rate, implantation rate, and miscarriage rate were compared between the groups. RESULTS Similar rates of fertilization (70% and 73%), cleavage (98% and 100%), and aneuploidy (76.9% and 70.9%) were observed in the IMSI and ICSI groups, respectively. The IMSI group had significantly more good quality embryos at day 3 (95% vs 73%), higher blastocyst development rates (33% vs 19%), and greater number of hatching blastocysts (43% vs 28%), cycles with at least one blastocyst at day 5 (55% vs 35%), and blastocysts with good trophoectoderm morphology (21% vs 6%) compared with the ICSI group (P < 0.001). Significantly higher implantation rates were observed in the IMSI group compared with the ICSI group (57% vs 27%; P < 0.05). Pregnancy and miscarriage rates were similar in both groups (80% vs 50% and 0% vs 33%, respectively). CONCLUSION The IMSI procedure significantly improves the embryo quality/development by increasing the implantation rates without affecting the chromosomal status of embryos. There is a tendency for the IMSI procedure to enhance the pregnancy rates and lower the miscarriage rates when compared with ICSI.
Collapse
Affiliation(s)
- Daniel Luna
- FERTILAB Laboratory of Assisted Reproduction, Lima, Perú
| | | | | | | | | | - Lucy Villegas
- FERTILAB Laboratory of Assisted Reproduction, Lima, Perú
| | | |
Collapse
|