1
|
Kakourou G, Sofocleous C, Mamas T, Vrettou C, Traeger-Synodinos J. The current clinical applications of preimplantation genetic testing (PGT): acknowledging the limitations of biology and technology. Expert Rev Mol Diagn 2024; 24:767-775. [PMID: 39107971 DOI: 10.1080/14737159.2024.2390187] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2024] [Accepted: 08/05/2024] [Indexed: 08/13/2024]
Abstract
INTRODUCTION Preimplantation Genetic Testing (PGT) is a cutting-edge test used to detect genetic abnormalities in embryos fertilized through Medically Assisted Reproduction (MAR). PGT aims to ensure that embryos selected for transfer are free of specific genetic conditions or chromosome abnormalities, thereby reducing chances for unsuccessful MAR cycles, complicated pregnancies, and genetic diseases in future children. AREAS COVERED In PGT, genetics, embryology, and technology progress and evolve together. Biological and technological limitations are described and addressed to highlight complexity and knowledge constraints and draw attention to concerns regarding safety of procedures, clinical validity, and utility, extent of applications and overall ethical implications for future families and society. EXPERT OPINION Understanding the genetic basis of diseases along with advanced technologies applied in embryology and genetics contribute to faster, cost-effective, and more efficient PGT. Next Generation Sequencing-based techniques, enhanced by improved bioinformatics, are expected to upgrade diagnostic accuracy. Complicating findings such as mosaicism, mt-DNA variants, variants of unknown significance, or variants related to late-onset or polygenic diseases will however need further appraisal. Emphasis on monitoring such emerging data is crucial for evidence-based counseling while standardized protocols and guidelines are essential to ensure clinical value and respect of Ethical, Legal and Societal Issues.
Collapse
Affiliation(s)
- Georgia Kakourou
- Laboratory of Medical Genetics, Medical School, National and Kapodistrian University of Athens, "Aghia Sophia" Children's Hospital, Athens, Greece
| | - Christalena Sofocleous
- Laboratory of Medical Genetics, Medical School, National and Kapodistrian University of Athens, "Aghia Sophia" Children's Hospital, Athens, Greece
| | - Thalia Mamas
- Laboratory of Medical Genetics, Medical School, National and Kapodistrian University of Athens, "Aghia Sophia" Children's Hospital, Athens, Greece
| | - Christina Vrettou
- Laboratory of Medical Genetics, Medical School, National and Kapodistrian University of Athens, "Aghia Sophia" Children's Hospital, Athens, Greece
| | - Joanne Traeger-Synodinos
- Laboratory of Medical Genetics, Medical School, National and Kapodistrian University of Athens, "Aghia Sophia" Children's Hospital, Athens, Greece
| |
Collapse
|
2
|
Zhang X, Zheng PS. Mechanism of chromosomal mosaicism in preimplantation embryos and its effect on embryo development. J Assist Reprod Genet 2024; 41:1127-1141. [PMID: 38386118 PMCID: PMC11143108 DOI: 10.1007/s10815-024-03048-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Accepted: 01/30/2024] [Indexed: 02/23/2024] Open
Abstract
Aneuploidy is one of the main causes of miscarriage and in vitro fertilization failure. Mitotic abnormalities in preimplantation embryos are the main cause of mosaicism, which may be influenced by several endogenous factors such as relaxation of cell cycle control mechanisms, defects in chromosome cohesion, centrosome aberrations and abnormal spindle assembly, and DNA replication stress. In addition, incomplete trisomy rescue is a rare cause of mosaicism. However, there may be a self-correcting mechanism in mosaic embryos, which allows some mosaicisms to potentially develop into normal embryos. At present, it is difficult to accurately diagnose mosaicism using preimplantation genetic testing for aneuploidy. Therefore, in clinical practice, embryos diagnosed as mosaic should be considered comprehensively based on the specific situation of the patient.
Collapse
Affiliation(s)
- Xue Zhang
- Department of Reproductive Medicine, The First Affiliated Hospital, Xi'an Jiaotong University of Medical School, Xi'an, 710061, Shanxi, P.R. China
| | - Peng-Sheng Zheng
- Department of Reproductive Medicine, The First Affiliated Hospital, Xi'an Jiaotong University of Medical School, Xi'an, 710061, Shanxi, P.R. China.
- Section of Cancer Stem Cell Research, Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education of People's Republic of China, Xi'an, 710061, Shanxi, P.R. China.
| |
Collapse
|
3
|
De Martin H, Bonetti TCS, Nissel CAZ, Gomes AP, Fujii MG, Monteleone PAA. Association of early cleavage, morula compaction and blastocysts ploidy of IVF embryos cultured in a time-lapse system and biopsied for genetic test for aneuploidy. Sci Rep 2024; 14:739. [PMID: 38185698 PMCID: PMC10772106 DOI: 10.1038/s41598-023-51087-z] [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: 06/01/2023] [Accepted: 12/30/2023] [Indexed: 01/09/2024] Open
Abstract
IVF embryos have historically been evaluated by morphological characteristics. The time-lapse system (TLS) has become a promising tool, providing an uninterrupted evaluation of morphological and dynamic parameters of embryo development. Furthermore, TLS sheds light on unknown phenomena such as direct cleavage and incomplete morula compaction. We retrospectively analyzed the morphology (Gardner Score) and morphokinetics (KIDScore) of 835 blastocysts grown in a TLS incubator (Embryoscope+), which were biopsied for preimplantation genetic testing for aneuploidy (PGT-A). Only the embryos that reached the blastocyst stage were included in this study and time-lapse videos were retrospectively reanalysed. According to the pattern of initial cleavages and morula compaction, the embryos were classified as: normal (NC) or abnormal (AC) cleavage, and fully (FCM) or partially compacted (PCM) morulae. No difference was found in early cleavage types or morula compaction patterns between female age groups (< 38, 38-40 and > 40 yo). Most of NC embryos resulted in FCM (≅ 60%), while no embryos with AC resulted in FCM. Aneuploidy rate of AC-PCM group did not differ from that of NC-FCM group in women < 38 yo, but aneuploidy was significantly higher in AC-PCM compared to NC-FCM of women > 40 yo. However, the quality of embryos was lower in AC-PCM blastocysts in women of all age ranges. Morphological and morphokinetic scores declined with increasing age, in the NC-PCM and AC-PCM groups, compared to the NC-FCM. Similar aneuploidy rates among NC-FCM and AC-PCM groups support the hypothesis that PCM in anomalous-cleaved embryos can represent a potential correction mechanism, even though lower morphological/morphokinetic scores are seen on AC-PCM. Therefore, both morphological and morphokinetic assessment should consider these embryonic development phenomena.
Collapse
Affiliation(s)
- H De Martin
- Centro de Reprodução Humana Monteleone, Rua Lima Barros, 61 Jardim Paulista, São Paulo, SP, CEP 04503-030, Brazil.
- Disciplina de Ginecologia-Departamento de Obstetrícia e Ginecologia, Faculdade de Medicina da Universidade de São Paulo, Av. Dr. Enéas Carvalho de Aguiar, 255-10 Andar-Cerqueira César, São Paulo, SP, CEP 05403-000, Brazil.
| | - T C S Bonetti
- Centro de Reprodução Humana Monteleone, Rua Lima Barros, 61 Jardim Paulista, São Paulo, SP, CEP 04503-030, Brazil
- Departamento de Ginecologia, Escola Paulista de Medicina - Universidade Federal de São Paulo, Rua Pedro de Toledo, 781. 4º andar. Vila Clementino, São Paulo, SP, 04039030, Brazil
| | - C A Z Nissel
- Centro de Reprodução Humana Monteleone, Rua Lima Barros, 61 Jardim Paulista, São Paulo, SP, CEP 04503-030, Brazil
- Disciplina de Ginecologia-Departamento de Obstetrícia e Ginecologia, Faculdade de Medicina da Universidade de São Paulo, Av. Dr. Enéas Carvalho de Aguiar, 255-10 Andar-Cerqueira César, São Paulo, SP, CEP 05403-000, Brazil
| | - A P Gomes
- Centro de Reprodução Humana Monteleone, Rua Lima Barros, 61 Jardim Paulista, São Paulo, SP, CEP 04503-030, Brazil
| | - M G Fujii
- Centro de Reprodução Humana Monteleone, Rua Lima Barros, 61 Jardim Paulista, São Paulo, SP, CEP 04503-030, Brazil
| | - P A A Monteleone
- Centro de Reprodução Humana Monteleone, Rua Lima Barros, 61 Jardim Paulista, São Paulo, SP, CEP 04503-030, Brazil
- Disciplina de Ginecologia-Departamento de Obstetrícia e Ginecologia, Faculdade de Medicina da Universidade de São Paulo, Av. Dr. Enéas Carvalho de Aguiar, 255-10 Andar-Cerqueira César, São Paulo, SP, CEP 05403-000, Brazil
| |
Collapse
|
4
|
Wei Y, Wang J, Qu R, Zhang W, Tan Y, Sha Y, Li L, Yin T. Genetic mechanisms of fertilization failure and early embryonic arrest: a comprehensive review. Hum Reprod Update 2024; 30:48-80. [PMID: 37758324 DOI: 10.1093/humupd/dmad026] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Revised: 07/07/2023] [Indexed: 10/03/2023] Open
Abstract
BACKGROUND Infertility and pregnancy loss are longstanding problems. Successful fertilization and high-quality embryos are prerequisites for an ongoing pregnancy. Studies have proven that every stage in the human reproductive process is regulated by multiple genes and any problem, at any step, may lead to fertilization failure (FF) or early embryonic arrest (EEA). Doctors can diagnose the pathogenic factors involved in FF and EEA by using genetic methods. With the progress in the development of new genetic technologies, such as single-cell RNA analysis and whole-exome sequencing, a new approach has opened up for us to directly study human germ cells and reproductive development. These findings will help us to identify the unique mechanism(s) that leads to FF and EEA in order to find potential treatments. OBJECTIVE AND RATIONALE The goal of this review is to compile current genetic knowledge related to FF and EEA, clarifying the mechanisms involved and providing clues for clinical diagnosis and treatment. SEARCH METHODS PubMed was used to search for relevant research articles and reviews, primarily focusing on English-language publications from January 1978 to June 2023. The search terms included fertilization failure, early embryonic arrest, genetic, epigenetic, whole-exome sequencing, DNA methylation, chromosome, non-coding RNA, and other related keywords. Additional studies were identified by searching reference lists. This review primarily focuses on research conducted in humans. However, it also incorporates relevant data from animal models when applicable. The results were presented descriptively, and individual study quality was not assessed. OUTCOMES A total of 233 relevant articles were included in the final review, from 3925 records identified initially. The review provides an overview of genetic factors and mechanisms involved in the human reproductive process. The genetic mutations and other genetic mechanisms of FF and EEA were systematically reviewed, for example, globozoospermia, oocyte activation failure, maternal effect gene mutations, zygotic genome activation abnormalities, chromosome abnormalities, and epigenetic abnormalities. Additionally, the review summarizes progress in treatments for different gene defects, offering new insights for clinical diagnosis and treatment. WIDER IMPLICATIONS The information provided in this review will facilitate the development of more accurate molecular screening tools for diagnosing infertility using genetic markers and networks in human reproductive development. The findings will also help guide clinical practice by identifying appropriate interventions based on specific gene mutations. For example, when an individual has obvious gene mutations related to FF, ICSI is recommended instead of IVF. However, in the case of genetic defects such as phospholipase C zeta1 (PLCZ1), actin-like7A (ACTL7A), actin-like 9 (ACTL9), and IQ motif-containing N (IQCN), ICSI may also fail to fertilize. We can consider artificial oocyte activation technology with ICSI to improve fertilization rate and reduce monetary and time costs. In the future, fertility is expected to be improved or restored by interfering with or supplementing the relevant genes.
Collapse
Affiliation(s)
- Yiqiu Wei
- Reproductive Medical Center, Renmin Hospital of Wuhan University, Wuhan, China
| | - Jingxuan Wang
- Reproductive Medical Center, Renmin Hospital of Wuhan University, Wuhan, China
| | - Rui Qu
- Reproductive Medical Center, Renmin Hospital of Wuhan University, Wuhan, China
| | - Weiqian Zhang
- Reproductive Medical Center, Renmin Hospital of Wuhan University, Wuhan, China
| | - Yiling Tan
- Reproductive Medical Center, Renmin Hospital of Wuhan University, Wuhan, China
| | - Yanwei Sha
- Department of Andrology, Women and Children's Hospital, School of Medicine, Xiamen University, Xiamen, China
- Fujian Provincial Key Laboratory of Reproductive Health Research, School of Medicine, Xiamen University, Xiamen, China
| | - Lin Li
- Central Laboratory, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing, China
| | - Tailang Yin
- Reproductive Medical Center, Renmin Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
5
|
Ezoe K, Takahashi T, Miki T, Kato K. Developmental perturbation in human embryos: Clinical and biological significance learned from time-lapse images. Reprod Med Biol 2024; 23:e12593. [PMID: 38983691 PMCID: PMC11232294 DOI: 10.1002/rmb2.12593] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Revised: 05/30/2024] [Accepted: 06/07/2024] [Indexed: 07/11/2024] Open
Abstract
Background Time-lapse technology (TLT) has gained widespread adoption worldwide. In addition to facilitating the undisturbed culture of embryos, TLT offers the unique capability of continuously monitoring embryos to detect spatiotemporal changes. Although these observed phenomena play a role in optimal embryo selection/deselection, the clinical advantages of introducing TLT remain unclear. However, manual annotation of embryo perturbation could facilitate a comprehensive assessment of developmental competence. This process requires a thorough understanding of embryo observation and the biological significance associated with developmental dogma and variation. This review elucidates the typical behavior and variation of each phenomenon, exploring their clinical significance and research perspectives. Methods The MEDLINE database was searched using PubMed for peer-reviewed English-language original articles concerning human embryo development. Main findings TLT allows the observation of consecutive changes in embryo morphology, serving as potential biomarkers for embryo assessment. In assisted reproductive technology laboratories, several phenomena have not revealed their mechanism, posing difficulties such as fertilization deficiency and morula arrest. Conclusion A profound understanding of the biological mechanisms and significance of each phenomenon is crucial. Further collaborative efforts between the clinical and molecular fields following translational studies are required to advance embryonic outcomes and assessment.
Collapse
|
6
|
Zhao H, Lv N, Cong J, Chen G, Bao H, Liu X. Upregulated RPA2 in endometrial tissues of repeated implantation failure patients impairs the endometrial decidualization. J Assist Reprod Genet 2023; 40:2739-2750. [PMID: 37831348 PMCID: PMC10643753 DOI: 10.1007/s10815-023-02946-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Accepted: 09/09/2023] [Indexed: 10/14/2023] Open
Abstract
PURPOSE To investigate the expression and underlying mechanism of RPA2 in endometrium of patients with repeated implantation failure (RIF). METHODS In this study, we retrieved the expression profiles from GEO databases and filtered the differentially expressed genes between RIF and the fertile control group. Ultimately, RPA2 was confirmed as a target gene. RPA2 expression in endometrial tissues of RIF patients, the control group, and different phases was detected by RT-qPCR, immunohistochemistry, and Western blotting. The role of RPA2 in endometrial decidualization was performed by in vitro decidualization inducing by 8-Br-cAMP and MPA. Furthermore, RT-qPCR was used to detect changes in the decidual biomarkers after transfection of RPA2 overexpression vector in human endometrium stromal cell (HESC). RESULTS RPA2 was significantly upregulated in the mid-secretory endometrium of patients with RIF. As a proliferation-related gene, RPA2 was obviously higher expressed at proliferative phase during the normal menstrual cycles. Moreover, the downregulation of RPA2 was discovered during decidualization of HESC. Furthermore, RPA2 overexpression impaired decidualization by inhibiting the expression of prolactin (PRL) and insulin-like growth factor-binding protein 1 (IGFBP1). CONCLUSIONS Our finding indicated that aberrant upregulation of RPA2 attenuated decidualization of HESC in RIF women and provided new potential therapeutic targets.
Collapse
Affiliation(s)
- Huishan Zhao
- Reproductive Medicine Centre, the Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, China
| | - Na Lv
- Department of Clinical Laboratory, the Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, China
| | - Jianxiang Cong
- Reproductive Medicine Centre, the Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, China
| | - Gang Chen
- Department of Breast Surgery, the Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, China
| | - Hongchu Bao
- Reproductive Medicine Centre, the Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, China.
| | - Xuemei Liu
- Reproductive Medicine Centre, the Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, China.
| |
Collapse
|
7
|
Watanabe S, Yoshikai K, Matsuda Y, Miyai S, Sawada Y, Kurahashi H, Sawada T. The effect of early irregular cell division of human embryos on blastocyst euploidy: considerations from the subsequent development of the blastomeres by direct or reverse cleavage. F&S SCIENCE 2023; 4:21-29. [PMID: 36410651 DOI: 10.1016/j.xfss.2022.11.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Revised: 11/11/2022] [Accepted: 11/15/2022] [Indexed: 11/21/2022]
Abstract
OBJECTIVE To investigate whether blastocysts that divide irregularly reduce subsequent blastocyst euploidy. DESIGN Retrospective study. SETTING Private clinic. PATIENT(S) A total of 122 blastocysts for which consent for disposal and research use was obtained. INTERVENTION(S) None. MAIN OUTCOME MEASURE(S) Results of next-generation sequencing analysis of the blastocysts and whether blastomeres by normal or irregular divisions subsequently participated in blastocyst formation or not. RESULT(S) The embryos were classified according to their dynamics until the second cleavage. The blastocyst euploidy rates were 33.3% (19/57) in the normal cleavage (NC) group, 38.3% (18/47) in the direct cleavage (embryos with one cell dividing into 3 cells) (DC) group, and 72.2% (13/18) in the reverse cleavage (RC) (embryos with fused cells once divided) group. The rate of the RC group was significantly higher than that of the NC group. The blastocyst participation rate of the blastomeres were 95.6% in the NC group and 56.5% in that derived from DC of the first cleavage, and 91.7% in that of blastomeres derived from normal division of the second cleavage and 53.6% in that derived from DC of the second cleavage, both of which were significantly lower in the latter. In the RC group, the rates of fused and nonfused blastomeres were 62.1% and 87.5%, respectively, with no significant difference. CONCLUSION(S) The blastomeres generated by DC were often excluded from blastocyst formation, and we speculate that this is one reason why their division does not reduce blastocyst euploidy. The association between RC and euploidy of blastocysts merits further study.
Collapse
Affiliation(s)
| | | | | | - Shunsuke Miyai
- Institute for Comprehensive Medical Science, Fujita Health University, Toyoake, Japan
| | - Yuki Sawada
- Sawada Women's Clinic, Nagoya, Japan; Department of Obstetrics and Gynecology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Hiroki Kurahashi
- Institute for Comprehensive Medical Science, Fujita Health University, Toyoake, Japan
| | | |
Collapse
|
8
|
Greco E, Yakovlev P, Kornilov N, Vyatkina S, Bogdanova D, Ermakova M, Tarasova Y, Tikhonov A, Pendina A, Biricik A, Sessa MT, Listorti I, Ronsini C, Greco PF, Victor A, Barnes F, Zouves C, Spinella F, Viotti M. Two clinical case reports of embryonic mosaicism identified with PGT-A persisting during pregnancy as true fetal mosaicism. Hum Reprod 2023; 38:315-323. [PMID: 36610460 DOI: 10.1093/humrep/deac263] [Citation(s) in RCA: 18] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2022] [Revised: 10/31/2022] [Indexed: 01/09/2023] Open
Abstract
The health risks associated with transferring embryos classified as mosaic by preimplantation genetic testing for aneuploidies (PGT-A) are currently unknown. Such embryos produce PGT-A results indicating the presence of both euploid and aneuploid cells and have historically been deselected from transfer and grouped with uniformly aneuploid embryos as 'abnormal'. In recent years, numerous groups have reported the intentional transfer of mosaic embryos in the absence of uniformly euploid embryos, largely observing births of seemingly healthy babies. However, it remains to be understood whether the embryonic mosaicism invariably becomes resolved during the ensuing pregnancy, or whether the placenta and/or fetal tissues retain aneuploid cells, and if so to what potential clinical effect. Here, we report two cases of mosaicism persisting from the embryonic stage to the established pregnancy. Case 1 involved an embryonic low-level segmental mosaic loss in Chromosome (Chr) 1, which was confirmed in amniocentesis as well as in brain tissue of the products of conception. This pregnancy was terminated due to the chromosomal pathologies associated with 1p36 deletion syndrome, such as severe intellectual disability. Case 2 involved a low-level mosaic Chr 21 trisomy, which was confirmed with chorionic villus sampling and amniocentesis. The ensuing pregnancy was terminated after ultrasound identification of severe abnormalities in the placenta and fetus. Together, these two cases should be taken into account for risk-benefit assessments of prospective mosaic embryo transfers.
Collapse
Affiliation(s)
- Ermanno Greco
- Villa Mafalda, Centre For Reproductive Medicine, Rome, Italy.,Department of Obstetrics and Gynecology, UniCamillus International University, Rome, Italy
| | - Pavel Yakovlev
- Next Generation Clinic, Centre For Reproductive Medicine, Moscow, Russia
| | - Nikolay Kornilov
- Next Generation Clinic, Centre For Reproductive Medicine, Moscow, Russia.,Next Generation Clinic, Centre For Reproductive Medicine, St. Petersburg, Russia
| | - Svetlana Vyatkina
- Next Generation Clinic, Centre For Reproductive Medicine, St. Petersburg, Russia
| | - Daria Bogdanova
- Next Generation Clinic, Centre For Reproductive Medicine, Moscow, Russia
| | - Marina Ermakova
- Medical Genetic Center of the Group of companies "Mother and Child", Moscow, Russia
| | - Yulia Tarasova
- Medical Genetic Center of the Group of companies "Mother and Child", Moscow, Russia
| | - Andrei Tikhonov
- D.O. Ott Research Institute of Obstetrics, Gynecology and Reproduction, Saint Petersburg, Russia
| | - Anna Pendina
- D.O. Ott Research Institute of Obstetrics, Gynecology and Reproduction, Saint Petersburg, Russia
| | - Anil Biricik
- Eurofins Genoma Group, Molecular Genetics Laboratories, Rome, Italy
| | | | - Ilaria Listorti
- Villa Mafalda, Centre For Reproductive Medicine, Rome, Italy
| | - Carlo Ronsini
- Dipartimento della Donna, del Bambino e di Chirurgia Generale e Specialistica, University of Campania Luigi Vanvitelli, Naples, Italy
| | | | | | | | | | | | - Manuel Viotti
- Zouves Fertility Center, Foster City, CA, USA.,Zouves Foundation for Reproductive Medicine, Foster City, CA, USA
| |
Collapse
|
9
|
Currie CE, Ford E, Benham Whyte L, Taylor DM, Mihalas BP, Erent M, Marston AL, Hartshorne GM, McAinsh AD. The first mitotic division of human embryos is highly error prone. Nat Commun 2022; 13:6755. [PMID: 36347869 PMCID: PMC9643329 DOI: 10.1038/s41467-022-34294-6] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Accepted: 10/20/2022] [Indexed: 11/09/2022] Open
Abstract
Human beings are made of ~50 trillion cells which arise from serial mitotic divisions of a single cell - the fertilised egg. Remarkably, the early human embryo is often chromosomally abnormal, and many are mosaic, with the karyotype differing from one cell to another. Mosaicism presumably arises from chromosome segregation errors during the early mitotic divisions, although these events have never been visualised in living human embryos. Here, we establish live cell imaging of chromosome segregation using normally fertilised embryos from an egg-share-to-research programme, as well as embryos deselected during fertility treatment. We reveal that the first mitotic division has an extended prometaphase/metaphase and exhibits phenotypes that can cause nondisjunction. These included multipolar chromosome segregations and lagging chromosomes that lead to formation of micronuclei. Analysis of nuclear number and size provides evidence of equivalent phenotypes in 2-cell human embryos that gave rise to live births. Together this shows that errors in the first mitotic division can be tolerated in human embryos and uncovers cell biological events that contribute to preimplantation mosaicism.
Collapse
Affiliation(s)
- Cerys E. Currie
- grid.7372.10000 0000 8809 1613Division of Biomedical Sciences, Warwick Medical School, University of Warwick, Coventry, CV4 7AL UK ,grid.7372.10000 0000 8809 1613Centre for Mechanochemical Cell Biology, University of Warwick, Coventry, CV4 7AL UK
| | - Emma Ford
- grid.7372.10000 0000 8809 1613Division of Biomedical Sciences, Warwick Medical School, University of Warwick, Coventry, CV4 7AL UK ,grid.7372.10000 0000 8809 1613Centre for Mechanochemical Cell Biology, University of Warwick, Coventry, CV4 7AL UK
| | - Lucy Benham Whyte
- grid.15628.380000 0004 0393 1193University Hospitals Coventry and Warwickshire NHS Trust, Coventry, CV2 2DX UK ,Present Address: Kings Fertility Ltd, Fetal Medicine Research Institute, 16-20 Windsor Walk, SE5 8SS London, UK
| | - Deborah M. Taylor
- grid.15628.380000 0004 0393 1193University Hospitals Coventry and Warwickshire NHS Trust, Coventry, CV2 2DX UK
| | - Bettina P. Mihalas
- grid.4305.20000 0004 1936 7988Wellcome Centre for Cell Biology, University of Edinburgh, Edinburgh, UK
| | - Muriel Erent
- grid.7372.10000 0000 8809 1613Division of Biomedical Sciences, Warwick Medical School, University of Warwick, Coventry, CV4 7AL UK ,grid.7372.10000 0000 8809 1613Centre for Mechanochemical Cell Biology, University of Warwick, Coventry, CV4 7AL UK
| | - Adele L. Marston
- grid.4305.20000 0004 1936 7988Wellcome Centre for Cell Biology, University of Edinburgh, Edinburgh, UK
| | - Geraldine M. Hartshorne
- grid.7372.10000 0000 8809 1613Division of Biomedical Sciences, Warwick Medical School, University of Warwick, Coventry, CV4 7AL UK ,grid.15628.380000 0004 0393 1193University Hospitals Coventry and Warwickshire NHS Trust, Coventry, CV2 2DX UK ,grid.7372.10000 0000 8809 1613Centre for Early Life, University of Warwick, Coventry, CV4 7AL UK
| | - Andrew D. McAinsh
- grid.7372.10000 0000 8809 1613Division of Biomedical Sciences, Warwick Medical School, University of Warwick, Coventry, CV4 7AL UK ,grid.7372.10000 0000 8809 1613Centre for Mechanochemical Cell Biology, University of Warwick, Coventry, CV4 7AL UK ,grid.7372.10000 0000 8809 1613Centre for Early Life, University of Warwick, Coventry, CV4 7AL UK
| |
Collapse
|
10
|
Albertini DF. The fan base for embryo selection strategies: enlightened or flummoxed. J Assist Reprod Genet 2022; 39:2437-2438. [PMID: 36447080 PMCID: PMC9723044 DOI: 10.1007/s10815-022-02672-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/03/2022] Open
|
11
|
Clinical Outcome, Socioeconomic Status and Psychological Constrains of Patients Undergoing Preimplantation Genetic Testing (PGT) in Northern Greece. Medicina (B Aires) 2022; 58:medicina58101493. [PMID: 36295653 PMCID: PMC9611400 DOI: 10.3390/medicina58101493] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2022] [Revised: 09/07/2022] [Accepted: 10/11/2022] [Indexed: 11/05/2022] Open
Abstract
Background and objectives: Preimplantation genetic testing (PGT) offers patients the possibility of having a healthy baby free of chromosomal or genetic disorders. The present study focuses on the application of PGT for patients located in Northern Greece, investigating their clinical outcomes, their motives, and their overall physical and emotional experience during the treatment, in association with their socioeconomic background. Materials and Methods: Couples who underwent PGT for a monogenic condition (PGT-M, n = 19 cycles) or aneuploidy (PGT-A, n = 22 cycles) participated in the study. Fertilization, implantation, and pregnancy rates were recorded for all cycles. The couples were asked to fill in a questionnaire about the consultation they had received prior to treatment, their sociodemographic information, and the psychological impact PGT had on both the female and male partner. Results: The fertilization, implantation, and ongoing pregnancy rates for the PGT-M and PGT-A cycles were 81.3%, 70.6%, and 52.9%, and 78.2%, 64.3%, and 57.1%, respectively. Females experienced more intense physical pain than their male partners while psychological pain was encountered by both partners and occasionally in higher instances in males. No typical socioeconomic background of the patients referred for PGT in Northern Greece was noticed. Conclusion: PGT is an attractive alternative to prenatal diagnosis (PND), aiming to establisha healthy pregnancy by identifying and avoiding the transfer of chromosomally or genetically abnormal embryos to the uterus. Although the benefits of PGT were well-received by all patients undergoing the procedure, psychological pain was evident and especially prominent in patients with a previous affected child or no normal embryos for transfer. Holistic counseling is of utmost importance in order to make patients' experience during their journey to have a healthy baby less emotionally demanding and help them make the right choices for the future.
Collapse
|
12
|
De Coster T, Masset H, Tšuiko O, Catteeuw M, Zhao Y, Dierckxsens N, Aparicio AL, Dimitriadou E, Debrock S, Peeraer K, de Ruijter-Villani M, Smits K, Van Soom A, Vermeesch JR. Parental genomes segregate into distinct blastomeres during multipolar zygotic divisions leading to mixoploid and chimeric blastocysts. Genome Biol 2022; 23:201. [PMID: 36184650 PMCID: PMC9528162 DOI: 10.1186/s13059-022-02763-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Accepted: 08/31/2022] [Indexed: 11/23/2022] Open
Abstract
BACKGROUND During normal zygotic division, two haploid parental genomes replicate, unite and segregate into two biparental diploid blastomeres. RESULTS Contrary to this fundamental biological tenet, we demonstrate here that parental genomes can segregate to distinct blastomeres during the zygotic division resulting in haploid or uniparental diploid and polyploid cells, a phenomenon coined heterogoneic division. By mapping the genomic landscape of 82 blastomeres from 25 bovine zygotes, we show that multipolar zygotic division is a tell-tale of whole-genome segregation errors. Based on the haplotypes and live-imaging of zygotic divisions, we demonstrate that various combinations of androgenetic, gynogenetic, diploid, and polyploid blastomeres arise via distinct parental genome segregation errors including the formation of additional paternal, private parental, or tripolar spindles, or by extrusion of paternal genomes. Hence, we provide evidence that private parental spindles, if failing to congress before anaphase, can lead to whole-genome segregation errors. In addition, anuclear blastomeres are common, indicating that cytokinesis can be uncoupled from karyokinesis. Dissociation of blastocyst-stage embryos further demonstrates that whole-genome segregation errors might lead to mixoploid or chimeric development in both human and cow. Yet, following multipolar zygotic division, fewer embryos reach the blastocyst stage and diploidization occurs frequently indicating that alternatively, blastomeres with genome-wide errors resulting from whole-genome segregation errors can be selected against or contribute to embryonic arrest. CONCLUSIONS Heterogoneic zygotic division provides an overarching paradigm for the development of mixoploid and chimeric individuals and moles and can be an important cause of embryonic and fetal arrest following natural conception or IVF.
Collapse
Affiliation(s)
- Tine De Coster
- Laboratory for Cytogenetics and Genome Research, Department of Human Genetics, KU Leuven, 3000, Leuven, Belgium
- Reproductive Biology Unit, Department of Internal Medicine, Reproduction and Population Medicine, Ghent University, 9820, Merelbeke, Belgium
| | - Heleen Masset
- Laboratory for Cytogenetics and Genome Research, Department of Human Genetics, KU Leuven, 3000, Leuven, Belgium
| | - Olga Tšuiko
- Laboratory for Cytogenetics and Genome Research, Department of Human Genetics, KU Leuven, 3000, Leuven, Belgium
| | - Maaike Catteeuw
- Reproductive Biology Unit, Department of Internal Medicine, Reproduction and Population Medicine, Ghent University, 9820, Merelbeke, Belgium
| | - Yan Zhao
- Laboratory for Cytogenetics and Genome Research, Department of Human Genetics, KU Leuven, 3000, Leuven, Belgium
| | - Nicolas Dierckxsens
- Laboratory for Cytogenetics and Genome Research, Department of Human Genetics, KU Leuven, 3000, Leuven, Belgium
| | - Ainhoa Larreategui Aparicio
- Department of Clinical Sciences, Faculty of Veterinary Medicine, Utrecht University, 3584CM, Utrecht, The Netherlands
- Hubrecht Institute, 3584CT, Utrecht, The Netherlands
| | - Eftychia Dimitriadou
- Laboratory for Cytogenetics and Genome Research, Department of Human Genetics, KU Leuven, 3000, Leuven, Belgium
| | - Sophie Debrock
- Leuven University Fertility Center, University Hospitals of Leuven, 3000, Leuven, Belgium
| | - Karen Peeraer
- Leuven University Fertility Center, University Hospitals of Leuven, 3000, Leuven, Belgium
| | - Marta de Ruijter-Villani
- Department of Clinical Sciences, Faculty of Veterinary Medicine, Utrecht University, 3584CM, Utrecht, The Netherlands
- Hubrecht Institute, 3584CT, Utrecht, The Netherlands
- Division of Woman and Baby, Department Obstetrics and Gynaecology, University Medical Centre Utrecht, 3508, GA, Utrecht, The Netherlands
| | - Katrien Smits
- Reproductive Biology Unit, Department of Internal Medicine, Reproduction and Population Medicine, Ghent University, 9820, Merelbeke, Belgium
| | - Ann Van Soom
- Reproductive Biology Unit, Department of Internal Medicine, Reproduction and Population Medicine, Ghent University, 9820, Merelbeke, Belgium
| | - Joris Robert Vermeesch
- Laboratory for Cytogenetics and Genome Research, Department of Human Genetics, KU Leuven, 3000, Leuven, Belgium.
| |
Collapse
|
13
|
Bamford T, Barrie A, Montgomery S, Dhillon-Smith R, Campbell A, Easter C, Coomarasamy A. Morphological and morphokinetic associations with aneuploidy: a systematic review and meta-analysis. Hum Reprod Update 2022; 28:656-686. [PMID: 35613016 DOI: 10.1093/humupd/dmac022] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Revised: 02/10/2022] [Indexed: 08/17/2023] Open
Abstract
BACKGROUND A time lapse system (TLS) is utilized in some fertility clinics with the aim of predicting embryo viability and chance of live birth during IVF. It has been hypothesized that aneuploid embryos display altered morphokinetics as a consequence of their abnormal chromosome complement. Since aneuploidy is one of the fundamental reasons for IVF failure and miscarriage, attention has focused on utilizing morphokinetics to develop models to non-invasively risk stratify embryos for ploidy status. This could avoid or reduce the costs associated with pre-implantation genetic testing for aneuploidy (PGT-A). Furthermore, TLS have provided an understanding of the true prevalence of other dysmorphisms. Hypothetically, the incorporation of morphological features into a model could act synergistically, improving a model's discriminative ability to predict ploidy status. OBJECTIVE AND RATIONALE The aim of this systematic review and meta-analysis was to investigate associations between ploidy status and morphokinetic or morphological features commonly denoted on a TLS. This will determine the feasibility of a prediction model for euploidy and summarize the most useful prognostic markers to be included in model development. SEARCH METHODS Five separate searches were conducted in Medline, Embase, PubMed and Cinahl from inception to 1 July 2021. Search terms and word variants included, among others, PGT-A, ploidy, morphokinetics and time lapse, and the latter were successively substituted for the following morphological parameters: fragmentation, multinucleation, abnormal cleavage and contraction. Studies were limited to human studies. OUTCOMES Overall, 58 studies were included incorporating over 40 000 embryos. All except one study had a moderate risk of bias in at least one domain when assessed by the quality in prognostic studies tool. Ten morphokinetic variables were significantly delayed in aneuploid embryos. When excluding studies using less reliable genetic technologies, the most notable variables were: time to eight cells (t8, 1.13 h, 95% CI: 0.21-2.05; three studies; n = 742; I2 = 0%), t9 (2.27 h, 95% CI: 0.5-4.03; two studies; n = 671; I2 = 33%), time to formation of a full blastocyst (tB, 1.99 h, 95% CI 0.15-3.81; four studies; n = 1640; I2 = 76%) and time to expanded blastocyst (tEB, 2.35 h, 95% CI: 0.06-4.63; four studies; n = 1640; I2 = 83%). There is potentially some prognostic potential in the degree of fragmentation, multinucleation persisting to the four-cell stage and frequency of embryo contractions. Reverse cleavage was associated with euploidy in this meta-analysis; however, this article argues that these are likely spurious results requiring further investigation. There was no association with direct unequal cleavage in an embryo that progressed to a blastocyst, or with multinucleation assessed on Day 2 or at the two-cell stage. However, owing to heterogeneous results and poor-quality evidence, associations between these morphological components needs to be investigated further before conclusions can be reliably drawn. WIDER IMPLICATIONS This first systematic review and meta-analysis of morphological and morphokinetic associations with ploidy status demonstrates the most useful morphokinetic variables, namely t8, t9 and tEB to be included in future model development. There is considerable variability within aneuploid and euploid embryos making definitively classifying them impossible; however, it is feasible that embryos could be prioritized for biopsy. Furthermore, these results support the mechanism by which algorithms for live birth may have predictive ability, suggesting aneuploidy causes delayed cytokinesis. We highlight significant heterogeneity in our results secondary to local conditions and diverse patient populations, therefore calling for future models to be robustly developed and tested in-house. If successful, such a model would constitute a meaningful breakthrough when accessing PGT-A is unsuitable for couples.
Collapse
Affiliation(s)
| | | | | | - Rima Dhillon-Smith
- Tommy's National Centre for Miscarriage Research, Institute of Metabolism and Systems Research, College of Medical and Dental Sciences, University of Birmingham, Edgbaston, UK
| | | | - Christina Easter
- Tommy's National Centre for Miscarriage Research, Institute of Metabolism and Systems Research, College of Medical and Dental Sciences, University of Birmingham, Edgbaston, UK
| | - Arri Coomarasamy
- Tommy's National Centre for Miscarriage Research, Institute of Metabolism and Systems Research, College of Medical and Dental Sciences, University of Birmingham, Edgbaston, UK
| |
Collapse
|
14
|
Coticchio G, Borini A, Zacà C, Makrakis E, Sfontouris I. Fertilization signatures as biomarkers of embryo quality. Hum Reprod 2022; 37:1704-1711. [PMID: 35640036 DOI: 10.1093/humrep/deac123] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Revised: 05/11/2022] [Indexed: 12/29/2022] Open
Abstract
Fertilization underpins the vital transition from gametic meiosis to embryonic mitosis. For decades, in human IVF, microscopic observation at a single time point has limited our appreciation of the morphokinetic complexity of this process. More recently, the introduction of time lapse technology-also enhanced by combination with artificial intelligence-has revealed the finest morphokinetic details of the beginning of human development. Overall, a picture has finally emerged in which the precise timing, morphology and geometry of several fertilization events offer clues to predict the fate of the embryo-a key aspect of assisted reproduction. In this scenario, correct unfolding of intra- and interpronuclear rearrangements emerge as a crucial factor to create a platform able to preserve genetic and cellular integrity at the first mitotic cleavage.
Collapse
|
15
|
Transmission ratio distortion of mutations in the master regulator of centriole biogenesis PLK4. Hum Genet 2022; 141:1785-1794. [PMID: 35536377 PMCID: PMC9556372 DOI: 10.1007/s00439-022-02461-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Accepted: 04/26/2022] [Indexed: 11/04/2022]
Abstract
The evolutionary conserved Polo-like kinase 4 (PLK4) is essential for centriole duplication, spindle assembly, and de novo centriole formation. In man, homozygous mutations in PLK4 lead to primary microcephaly, altered PLK4 expression is associated with aneuploidy in human embryos. Here, we report on a consanguineous four-generation family with 8 affected individuals compound heterozygous for a novel missense variant, c.881 T > G, and a deletion of the PLK4 gene. The clinical phenotype of the adult patients is mild compared to individuals with previously described PLK4 mutations. One individual was homozygous for the variant c.881G and phenotypically unaffected. The deletion was inherited by 14 of 16 offspring and thus exhibits transmission ratio distortion (TRD). Moreover, based on the already published families with PLK4 mutations, it could be shown that due to the preferential transmission of the mutant alleles, the number of affected offspring is significantly increased. It is assumed that reduced expression of PLK4 decreases the intrinsically high error rate of the first cell divisions after fertilization, increases the number of viable embryos and thus leads to preferential transmission of the deleted/mutated alleles.
Collapse
|
16
|
Brooks KE, Daughtry BL, Davis B, Yan MY, Fei SS, Shepherd S, Carbone L, Chavez SL. Molecular contribution to embryonic aneuploidy and karyotypic complexity in initial cleavage divisions of mammalian development. Development 2022; 149:dev198341. [PMID: 35311995 PMCID: PMC9058497 DOI: 10.1242/dev.198341] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Accepted: 03/04/2022] [Indexed: 01/05/2023]
Abstract
Embryonic aneuploidy is highly complex, often leading to developmental arrest, implantation failure or spontaneous miscarriage in both natural and assisted reproduction. Despite our knowledge of mitotic mis-segregation in somatic cells, the molecular pathways regulating chromosome fidelity during the error-prone cleavage-stage of mammalian embryogenesis remain largely undefined. Using bovine embryos and live-cell fluorescent imaging, we observed frequent micro-/multi-nucleation of mis-segregated chromosomes in initial mitotic divisions that underwent unilateral inheritance, re-fused with the primary nucleus or formed a chromatin bridge with neighboring cells. A correlation between a lack of syngamy, multipolar divisions and asymmetric genome partitioning was also revealed, and single-cell DNA-seq showed propagation of primarily non-reciprocal mitotic errors. Depletion of the mitotic checkpoint protein BUB1B (also known as BUBR1) resulted in similarly abnormal nuclear structures and cell divisions, as well as chaotic aneuploidy and dysregulation of the kinase-substrate network that mediates mitotic progression, all before zygotic genome activation. This demonstrates that embryonic micronuclei sustain multiple fates, provides an explanation for blastomeres with uniparental origins, and substantiates defective checkpoints and likely other maternally derived factors as major contributors to the karyotypic complexity afflicting mammalian preimplantation development.
Collapse
Affiliation(s)
- Kelsey E. Brooks
- Division of Reproductive and Developmental Sciences, Oregon National Primate Research Center, Beaverton, OR 97006, USA
| | - Brittany L. Daughtry
- Division of Reproductive and Developmental Sciences, Oregon National Primate Research Center, Beaverton, OR 97006, USA
- Department of Cell, Developmental and Cancer Biology, Oregon Health and Science University, Portland, OR 97239, USA
| | - Brett Davis
- Bioinformatics and Biostatistics Unit, Oregon National Primate Research Center, Beaverton, OR 97006, USA
- Department of Medicine, Knight Cardiovascular Institute, Oregon Health and Science University, Portland, OR 97239, USA
| | - Melissa Y. Yan
- Bioinformatics and Biostatistics Unit, Oregon National Primate Research Center, Beaverton, OR 97006, USA
| | - Suzanne S. Fei
- Bioinformatics and Biostatistics Unit, Oregon National Primate Research Center, Beaverton, OR 97006, USA
| | - Selma Shepherd
- Division of Reproductive and Developmental Sciences, Oregon National Primate Research Center, Beaverton, OR 97006, USA
| | - Lucia Carbone
- Department of Medicine, Knight Cardiovascular Institute, Oregon Health and Science University, Portland, OR 97239, USA
- Division of Genetics, Oregon National Primate Research Center, Beaverton, OR 97006, USA
- Department of Medical Informatics and Clinical Epidemiology, Division of Bioinformatics and Computational Biomedicine, Oregon Health and Science University, Portland, OR 97239, USA
- Department of Molecular and Medical Genetics, Oregon Health and Science University, Portland, OR 97239, USA
| | - Shawn L. Chavez
- Division of Reproductive and Developmental Sciences, Oregon National Primate Research Center, Beaverton, OR 97006, USA
- Department of Molecular and Medical Genetics, Oregon Health and Science University, Portland, OR 97239, USA
- Department of Obstetrics and Gynecology, Oregon Health and Science University, Portland, OR 97239, USA
- Department of Biomedical Engineering, Oregon Health and Science University, Portland, OR 97239, USA
| |
Collapse
|
17
|
West JD, Everett CA. Preimplantation chromosomal mosaics, chimaeras and confined placental mosaicism. REPRODUCTION AND FERTILITY 2022; 3:R66-R90. [PMID: 35514539 PMCID: PMC9066951 DOI: 10.1530/raf-21-0095] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Accepted: 04/05/2022] [Indexed: 02/06/2023] Open
Abstract
Some human preimplantation embryos are chromosomally mosaic. For technical reasons, estimates of the overall frequency vary widely from <15 to >90% and the true frequency remains unknown. Aneuploid/diploid and aneuploid/aneuploid mosaics typically arise during early cleavage stages before the embryonic genome is fully activated and when cell cycle checkpoints are not operating normally. Other mosaics include chaotic aneuploid mosaics and mixoploids, some of which arise by abnormal chromosome segregation at the first cleavage division. Chimaeras are similar to mosaics, in having two genetically distinct cell populations, but they arise from more than one zygote and occur less often. After implantation, the frequency of mosaic embryos declines to about 2% and most are trisomic/diploid mosaics, with trisomic cells confined to the placenta. Thus, few babies are born with chromosomal mosaicism. This review discusses the origin of different types of chromosomal mosaics and chimaeras; their fate and the relationship between preimplantation chromosomal mosaicism and confined placental mosaicism in human conceptuses and animal models. Abnormal cells in mosaic embryos may be depleted by cell death, other types of cell selection or cell correction but the most severely affected mosaic embryos probably die. Trisomic cells could become restricted to placental lineages if cell selection or correction is less effective in placental lineages and/or they are preferentially allocated to a placental lineage. However, the relationship between preimplantation mosaicism and confined placental mosaicism may be complex because the specific chromosome(s) involved will influence whether chromosomally abnormal cells survive predominately in the placental trophoblast and/or placental mesenchyme. Lay summary Human cells normally have 23 pairs of chromosomes, which carry the genes. During the first few days of development, some human embryos are chromosomal mosaics. These mosaic embryos have both normal cells and cells with an abnormal number of chromosomes, which arise from the same fertilised egg. (More rarely, the different cell populations arise from more than one fertilised egg and these embryos are called chimaeras.) If chromosomally abnormal cells survive to term, they could cause birth defects. However, few abnormal cells survive and those that do are usually confined to the placenta, where they are less likely to cause harm. It is not yet understood how this restriction occurs but the type of chromosomal abnormality influences which placental tissues are affected. This review discusses the origin of different types of chromosomally abnormal cells, their fate and how they might become confined to the placenta in humans and animal models.
Collapse
Affiliation(s)
- John D West
- Section of Obstetrics and Gynaecology, Clinical Sciences, The Queen's Medical Research Institute, The University of Edinburgh, Edinburgh, UK
| | - Clare A Everett
- Section of Obstetrics and Gynaecology, Clinical Sciences, The Queen's Medical Research Institute, The University of Edinburgh, Edinburgh, UK
| |
Collapse
|
18
|
Chatzimeletiou K, Petrogiannis N, Sioga A, Emmanouil-Nikoloussi EN, Panagiotidis Y, Prapa M, Patrikiou A, Filippa M, Zervakakou G, Papanikolaou K, Makedos A, Kolibianakis E, Tarlatzis BC, Grimbizis G. The human embryo following biopsy on day 5 vs day 3: viability, ultrastructure and spindle / chromosomes configurations. Reprod Biomed Online 2022; 45:219-233. [DOI: 10.1016/j.rbmo.2022.02.022] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Revised: 02/23/2022] [Accepted: 02/24/2022] [Indexed: 11/26/2022]
|
19
|
Ghevaria H, SenGupta S, Naja R, Odia R, Exeter H, Serhal P, Gonzalez XV, Sun X, Delhanty J. Next Generation Sequencing Detects Premeiotic Errors in Human Oocytes. Int J Mol Sci 2022; 23:ijms23020665. [PMID: 35054849 PMCID: PMC8776218 DOI: 10.3390/ijms23020665] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Revised: 12/24/2021] [Accepted: 01/04/2022] [Indexed: 02/06/2023] Open
Abstract
Autosomal aneuploidy is the leading cause of embryonic and foetal death in humans. This arises mainly from errors in meiosis I or II of oogenesis. A largely ignored source of error stems from germinal mosaicism, which leads to premeiotic aneuploidy. Molecular cytogenetic studies employing metaphase fluorescence in situ hybridization and comparative genomic hybridisation suggest that premeiotic aneuploidy may affect 10–20% of oocytes overall. Such studies have been criticised on technical grounds. We report here an independent study carried out on unmanipulated oocytes that have been analysed using next generation sequencing (NGS). This study confirms that the incidence of premeiotic aneuploidy in an unselected series of oocytes exceeds 10%. A total of 140 oocytes donated by 42 women gave conclusive results; of these, 124 (88.5%) were euploid. Sixteen out of 140 (11.4%) provided evidence of premeiotic aneuploidy. Of the 140, 112 oocytes were immature (germinal vesicle or metaphase I), of which 10 were aneuploid (8.93%); the remaining 28 were intact metaphase II - first polar body complexes, and six of these were aneuploid (21.4%). Of the 16 aneuploid cells, half contained simple errors (one or two abnormal chromosomes) and half contained complex errors. We conclude that germinal mosaicism leading to premeiotic aneuploidy is a consistent finding affecting at least 10% of unselected oocytes from women undergoing egg collection for a variety of reasons. The importance of premeiotic aneuploidy lies in the fact that, for individual oocytes, it greatly increases the risk of an aneuploid mature oocyte irrespective of maternal age. As such, this may account for some cases of aneuploid conceptions in very young women.
Collapse
Affiliation(s)
- Harita Ghevaria
- Preimplantation Genetics Group, Institute for Women’s Health, University College London (UCL), London WC1E 6HX, UK; (H.G.); (R.N.); (X.V.G.); (X.S.); (J.D.)
| | - Sioban SenGupta
- Preimplantation Genetics Group, Institute for Women’s Health, University College London (UCL), London WC1E 6HX, UK; (H.G.); (R.N.); (X.V.G.); (X.S.); (J.D.)
- Correspondence:
| | - Roy Naja
- Preimplantation Genetics Group, Institute for Women’s Health, University College London (UCL), London WC1E 6HX, UK; (H.G.); (R.N.); (X.V.G.); (X.S.); (J.D.)
| | - Rabi Odia
- Embryology Department, The Centre for Reproductive and Genetic Health, London W1W 5QS, UK; (R.O.); (H.E.)
| | - Holly Exeter
- Embryology Department, The Centre for Reproductive and Genetic Health, London W1W 5QS, UK; (R.O.); (H.E.)
| | - Paul Serhal
- Clinical Department, The Centre for Reproductive and Genetic Health, London W1W 5QS, UK;
| | - Xavier Viñals Gonzalez
- Preimplantation Genetics Group, Institute for Women’s Health, University College London (UCL), London WC1E 6HX, UK; (H.G.); (R.N.); (X.V.G.); (X.S.); (J.D.)
| | - Xuhui Sun
- Preimplantation Genetics Group, Institute for Women’s Health, University College London (UCL), London WC1E 6HX, UK; (H.G.); (R.N.); (X.V.G.); (X.S.); (J.D.)
| | - Joy Delhanty
- Preimplantation Genetics Group, Institute for Women’s Health, University College London (UCL), London WC1E 6HX, UK; (H.G.); (R.N.); (X.V.G.); (X.S.); (J.D.)
| |
Collapse
|
20
|
Haplotype-aware inference of human chromosome abnormalities. Proc Natl Acad Sci U S A 2021; 118:2109307118. [PMID: 34772814 DOI: 10.1073/pnas.2109307118] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/16/2021] [Indexed: 12/25/2022] Open
Abstract
Extra or missing chromosomes-a phenomenon termed aneuploidy-frequently arise during human meiosis and embryonic mitosis and are the leading cause of pregnancy loss, including in the context of in vitro fertilization (IVF). While meiotic aneuploidies affect all cells and are deleterious, mitotic errors generate mosaicism, which may be compatible with healthy live birth. Large-scale abnormalities such as triploidy and haploidy also contribute to adverse pregnancy outcomes, but remain hidden from standard sequencing-based approaches to preimplantation genetic testing for aneuploidy (PGT-A). The ability to reliably distinguish meiotic and mitotic aneuploidies, as well as abnormalities in genome-wide ploidy, may thus prove valuable for enhancing IVF outcomes. Here, we describe a statistical method for distinguishing these forms of aneuploidy based on analysis of low-coverage whole-genome sequencing data, which is the current standard in the field. Our approach overcomes the sparse nature of the data by leveraging allele frequencies and linkage disequilibrium (LD) measured in a population reference panel. The method, which we term LD-informed PGT-A (LD-PGTA), retains high accuracy down to coverage as low as 0.05 × and at higher coverage can also distinguish between meiosis I and meiosis II errors based on signatures spanning the centromeres. LD-PGTA provides fundamental insight into the origins of human chromosome abnormalities, as well as a practical tool with the potential to improve genetic testing during IVF.
Collapse
|
21
|
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: 21] [Impact Index Per Article: 7.0] [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
|
22
|
Tšuiko O, Vanneste M, Melotte C, Ding J, Debrock S, Masset H, Peters M, Salumets A, De Leener A, Pirard C, Kluyskens C, Hostens K, van de Vijver A, Peeraer K, Denayer E, Vermeesch JR, Dimitriadou E. Haplotyping-based preimplantation genetic testing reveals parent-of-origin specific mechanisms of aneuploidy formation. NPJ Genom Med 2021; 6:81. [PMID: 34620870 PMCID: PMC8497526 DOI: 10.1038/s41525-021-00246-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Accepted: 09/16/2021] [Indexed: 11/17/2022] Open
Abstract
Chromosome instability is inherent to human IVF embryos, but the full spectrum and developmental fate of chromosome anomalies remain uncharacterized. Using haplotyping-based preimplantation genetic testing for monogenic diseases (PGT-M), we mapped the parental and mechanistic origin of common and rare genomic abnormalities in 2300 cleavage stage and 361 trophectoderm biopsies. We show that while single whole chromosome aneuploidy arises due to chromosome-specific meiotic errors in the oocyte, segmental imbalances predominantly affect paternal chromosomes, implicating sperm DNA damage in segmental aneuploidy formation. We also show that postzygotic aneuploidy affects multiple chromosomes across the genome and does not discriminate between parental homologs. In addition, 6% of cleavage stage embryos demonstrated signatures of tripolar cell division with excessive chromosome loss, however hypodiploid blastomeres can be excluded from further embryo development. This observation supports the selective-pressure hypothesis in embryos. Finally, considering that ploidy violations may constitute a significant proportion of non-viable embryos, using haplotyping-based approach to map these events might further improve IVF success rate.
Collapse
Affiliation(s)
- Olga Tšuiko
- Department of Human Genetics, Centre for Human Genetics, University Hospitals Leuven, Leuven, 3000, Belgium.,Laboratory of Cytogenetics and Genome Research, Centre for Human Genetics, KU Leuven, Leuven, 3000, Belgium
| | - Michiel Vanneste
- Department of Human Genetics, Centre for Human Genetics, University Hospitals Leuven, Leuven, 3000, Belgium
| | - Cindy Melotte
- Department of Human Genetics, Centre for Human Genetics, University Hospitals Leuven, Leuven, 3000, Belgium
| | - Jia Ding
- Department of Human Genetics, Centre for Human Genetics, University Hospitals Leuven, Leuven, 3000, Belgium
| | - Sophie Debrock
- Leuven University Fertility Center, University Hospitals Leuven, Leuven, 3000, Belgium
| | - Heleen Masset
- Laboratory of Cytogenetics and Genome Research, Centre for Human Genetics, KU Leuven, Leuven, 3000, Belgium
| | - Maire Peters
- Department of Obstetrics and Gynaecology, Institute of Clinical Medicine, University of Tartu, Tartu, 50406, Estonia
| | - Andres Salumets
- Department of Obstetrics and Gynaecology, Institute of Clinical Medicine, University of Tartu, Tartu, 50406, Estonia
| | - Anne De Leener
- Centre for Human Genetics, Cliniques Universitaires Saint Luc, UCLouvain, Brussels, 1200, Belgium
| | - Céline Pirard
- Department of Gynaecology, Cliniques Universitaires Saint Luc, UCLouvain, Brussels, 1200, Belgium
| | - Candice Kluyskens
- Department of Gynaecology, Cliniques Universitaires Saint Luc, UCLouvain, Brussels, 1200, Belgium
| | - Katleen Hostens
- Centre for Reproductive Medicine (CRG)-Brugge-Kortrijk, AZ Sint-Jan Brugge-Oostende AV, Brugge, 8000, Belgium
| | - Arne van de Vijver
- Centre for Reproductive Medicine (CRG)-Brugge-Kortrijk, AZ Sint-Jan Brugge-Oostende AV, Brugge, 8000, Belgium
| | - Karen Peeraer
- Leuven University Fertility Center, University Hospitals Leuven, Leuven, 3000, Belgium
| | - Ellen Denayer
- Department of Human Genetics, Centre for Human Genetics, University Hospitals Leuven, Leuven, 3000, Belgium
| | - Joris Robert Vermeesch
- Department of Human Genetics, Centre for Human Genetics, University Hospitals Leuven, Leuven, 3000, Belgium. .,Laboratory of Cytogenetics and Genome Research, Centre for Human Genetics, KU Leuven, Leuven, 3000, Belgium.
| | - Eftychia Dimitriadou
- Department of Human Genetics, Centre for Human Genetics, University Hospitals Leuven, Leuven, 3000, Belgium.
| |
Collapse
|
23
|
Chatzimeletiou K, Sioga A, Petrogiannis N, Panagiotidis Y, Prapa M, Patrikiou A, Tarlatzis BC, Grimbizis G. Viability assessment using fluorescent markers and ultrastructure of human biopsied embryos vitrified in open and closed systems. Reprod Biomed Online 2021; 43:833-842. [PMID: 34593325 DOI: 10.1016/j.rbmo.2021.05.011] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2021] [Revised: 05/04/2021] [Accepted: 05/11/2021] [Indexed: 01/14/2023]
Abstract
RESEARCH QUESTION Are there any differences in viability and ultrastructure amongst embryos biopsied on Day 5 versus Day 3 following vitrification in open and closed systems and compared to fresh embryos? DESIGN One hundred human embryos (40 blastocysts biopsied on Day 5 and subsequently vitrified in open or closed systems and 60 Day 3 biopsied embryos that developed to blastocysts but were rejected for transfer following preimplantation genetic testing for monogenic/single gene defects and for aneuploidies were either treated fresh [n = 20] or vitrified [n = 40] in open or closed systems) and following warming and culture for 4 h were subjected to viability staining with carboxyfluorescein-diacetate succinimidylester/propidium iodide or processed for transmission electron microscopy. RESULTS No statistically significant differences were observed in the viability of human biopsied embryos following vitrification in open and closed systems. Compared to fresh embryos, vitrified ones had a higher incidence of damage (propidium iodide-stained cells) irrespective of the vitrification method (P = 0.005). These damaged cells were more prominent in Day 5 biopsied blastocysts and mainly located at the position of cutting. Characteristic lipofuscin droplets (representative of apoptosis) and a higher number of vacuoles and distension of mitochondria were also more evident in vitrified embryos, although this was not statistically assessed. CONCLUSIONS Vitrification in open and closed systems does not adversely affect the viability and ultrastructure of Day 5 and Day 3 biopsied embryos as revealed by the minimal yet statistically significant cell damage observed. This damage may be compensated by the embryos, which in their attempt to fully recover following vitrification, potentially enable 'rescue' processes to eliminate it.
Collapse
Affiliation(s)
- Katerina Chatzimeletiou
- Unit for Human Reproduction, 1st Department of Obstetrics and Gynecology, Aristotle University Medical School, Papageorgiou General Hospital, Thessaloniki 56403, Greece.
| | - Antonia Sioga
- Laboratory of Histology and Embryology, Aristotle University Medical School, Thessaloniki 54124, Greece
| | | | | | - Marialena Prapa
- Iakentro Advanced Medical Centre, Thessaloniki 54250, Greece
| | - Antonios Patrikiou
- Unit for Human Reproduction, 1st Department of Obstetrics and Gynecology, Aristotle University Medical School, Papageorgiou General Hospital, Thessaloniki 56403, Greece
| | - Basil C Tarlatzis
- Unit for Human Reproduction, 1st Department of Obstetrics and Gynecology, Aristotle University Medical School, Papageorgiou General Hospital, Thessaloniki 56403, Greece
| | - Grigoris Grimbizis
- Unit for Human Reproduction, 1st Department of Obstetrics and Gynecology, Aristotle University Medical School, Papageorgiou General Hospital, Thessaloniki 56403, Greece
| |
Collapse
|
24
|
Ferraretto X, Hammas K, Llabador MA, Gricourt S, Labrosse J, Lousqui J, Epelboin S, Tubiana S, Patrat C. Early embryo development anomalies identified by time-lapse system: prevalence and impacting factors. Reprod Biomed Online 2021; 43:627-636. [PMID: 34474978 DOI: 10.1016/j.rbmo.2021.06.010] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Revised: 05/17/2021] [Accepted: 06/11/2021] [Indexed: 11/19/2022]
Abstract
RESEARCH QUESTION What is the prevalence of embryo abnormal early cleavage (ACL) identified by time lapse and factors related to patients and treatment that explain ACL occurrence? DESIGN A single-centre, retrospective cohort study. Data were collected on all IVF cycles for which embryos were observed in the EmbryoScope® between December 2015 and August 2017. Only diploid zygotes cleaved on day 2 were included. The study included 318 cycles (250 couples and 1343 embryos). Embryo videos were retrospectively analysed for ACL. The prevalence of each type of ACL was recorded. The influence of clinical factors (whether they were intrinsic to patients or specific to IVF treatment) on ACL occurrence was analysed in multivariate multilevel mixed-effect logistic regression analysis. RESULTS A high prevalence of ACL was observed: 37.6% (505/1343) of embryos presented at least one ACL, 22.8% (306/1343) a trichotomous mitosis, 25.8% (347/1343) a rapid cleavage, 6.7% (90/1343) a cell fusion and two or more ACL (16.1%). Part of the variation (12-25%) in ACL occurrence could be explained by embryo origin. Trichotomous mitosis and two or more ACL phenotypes were less likely to occur in women with endometriosis or tubal pathology and tubal pathology alone, respectively. No factor related to IVF cycles was found to be statistically associated with ACL occurrence. CONCLUSIONS Our findings emphasize the importance of considering embryo origin when interpreting studies focusing on embryo characteristics and factors that could affect their quality. The present study is limited by a small sample size of known embryo implantations and monocentric criterion.
Collapse
Affiliation(s)
- Xavier Ferraretto
- Service de Biologie de la Reproduction, AP-HP.Nord - Université de Paris, Hôpital Bichat, Paris 75018, France
| | - Karima Hammas
- Departement d'Epidemiologie, Biostatistiques et Recherche Clinique, AP-HP.Nord -Université de Paris 75018, Hôpital Bichat, Paris, France; Inserm, CIC-EC 1425, Hôpital Bichat Claude Bernard, Paris 75018, France
| | - Marie-Astrid Llabador
- Service de Biologie de la Reproduction, AP-HP.Nord - Université de Paris, Hôpital Bichat, Paris 75018, France
| | - Solenne Gricourt
- Service de Gynécologie, Obstétrique et Reproduction, AP-HP.Nord - Université de Paris, Hôpital Bichat, Paris 75018, France
| | - Julie Labrosse
- Service de Gynécologie, Obstétrique et Reproduction, AP-HP.Nord - Université de Paris, Hôpital Bichat, Paris 75018, France
| | - Johanna Lousqui
- Service de Biologie de la Reproduction, AP-HP.Nord - Université de Paris, Hôpital Bichat, Paris 75018, France
| | - Sylvie Epelboin
- Service de Gynécologie, Obstétrique et Reproduction, AP-HP.Nord - Université de Paris, Hôpital Bichat, Paris 75018, France
| | - Sarah Tubiana
- Departement d'Epidemiologie, Biostatistiques et Recherche Clinique, AP-HP.Nord -Université de Paris 75018, Hôpital Bichat, Paris, France; Inserm, CIC-EC 1425, Hôpital Bichat Claude Bernard, Paris 75018, France
| | - Catherine Patrat
- Service de Biologie de la Reproduction-CECOS, AP-HP.Centre - Université de Paris, Hôpital Cochin, Paris 75014, France; Université de Paris, Institut Cochin, U1016-CNRS UMR8104, Paris 75014, France.
| |
Collapse
|
25
|
Cimadomo D, Capalbo A, Dovere L, Tacconi L, Soscia D, Giancani A, Scepi E, Maggiulli R, Vaiarelli A, Rienzi L, Ubaldi FM. Leave the past behind: women's reproductive history shows no association with blastocysts' euploidy and limited association with live birth rates after euploid embryo transfers. Hum Reprod 2021; 36:929-940. [PMID: 33608730 DOI: 10.1093/humrep/deab014] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2020] [Revised: 12/30/2020] [Indexed: 12/20/2022] Open
Abstract
STUDY QUESTION Is there an association between patients' reproductive history and the mean euploidy rates per biopsied blastocysts (m-ER) or the live birth rates (LBRs) per first single vitrified-warmed euploid blastocyst transfers? SUMMARY ANSWER Patients' reproductive history (as annotated during counselling) showed no association with the m-ER, but a lower LBR was reported after euploid blastocyst transfer in women with a history of repeated implantation failure (RIF). WHAT IS KNOWN ALREADY Several studies have investigated the association between the m-ER and (i) patients' basal characteristics, (ii) ovarian stimulation strategy and dosage, (iii) culture media and conditions, and (iv) embryo morphology and day of full blastocyst development. Conversely, the expected m-ER due to women's reproductive history (previous live births (LBs), miscarriages, failed IVF cycles and transfers, and lack of euploid blastocysts among prior cohorts of biopsied embryos) still needs investigations. Yet, this information is critical to counsel new patients about a first cycle with preimplantation genetic testing for aneuploidy (PGT-A), but even more so after former adverse outcomes to prevent treatment drop-out. STUDY DESIGN, SIZE, DURATION This observational study included all patients undergoing a comprehensive chromosome testing (CCT)-based PGT-A cycle with at least one biopsied blastocyst in the period April 2013-December 2019 at a private IVF clinic (n = 2676 patients undergoing 2676 treatments and producing and 8151 blastocysts). m-ER were investigated according to women's reproductive history of LBs: no/≥1, miscarriages: no/1/>1; failed IVF cycles: no/1/2/>2, and implantation failures after previous transfers: no/1/2/>2. Among the 2676 patients included in this study, 440 (16%) had already undergone PGT-A before the study period; the data from these patients were further clustered according to the presence or absence of euploid embryo(s) in their previous cohort of biopsied blastocysts. The clinical outcomes per first single vitrified-warmed euploid blastocyst transfers (n =1580) were investigated according to the number of patients' previous miscarriages and implantation failures. PARTICIPANTS/MATERIALS, SETTING, METHODS The procedures involved in this study included ICSI, blastocyst culture, trophectoderm biopsy without hatching in Day 3, CCT-based PGT-A without reporting segmental and/or putative mitotic (or mosaic) aneuploidies and single vitrified-warmed euploid blastocyst transfer. For statistical analysis, Mann-Whitney U or Kruskal-Wallis tests, as well as linear regressions and generalised linear models among ranges of maternal age at oocyte retrieval were performed to identify significant differences for continuous variables. Fisher's exact tests and multivariate logistic regression analyses were instead used for categorical variables. MAIN RESULTS AND THE ROLE OF CHANCE Maternal age at oocyte retrieval was the only variable significantly associated with the m-ER. We defined five clusters (<35 years: 66 ± 31%; 35-37 years: 58 ± 33%; 38-40 years: 43 ± 35%; 40-42 years: 28 ± 34%; and >42 years: 17 ± 31%) and all analyses were conducted among them. The m-ER did not show any association with the number of previous LBs, miscarriages, failed IVF cycles or implantation failures. Among patients who had already undergone PGT-A before the study period, the m-ER did not associate with the absence (or presence) of euploid blastocysts in their former cohort of biopsied embryos. Regarding clinical outcomes of the first single vitrified-warmed euploid blastocyst transfer, the implantation rate was 51%, the miscarriage rate was 14% and the LBR was 44%. This LBR was independent of the number of previous miscarriages, but showed a decreasing trend depending on the number of previous implantation failures, reaching statistical significance when comparing patients with >2 failures and patients with no prior failure (36% versus 47%, P < 0.01; multivariate-OR adjusted for embryo quality and day of full blastocyst development: 0.64, 95% CI 0.48-0.86, P < 0.01). No such differences were shown for previous miscarriage rates. LIMITATIONS, REASONS FOR CAUTION The sample size for treatments following a former completed PGT-A cycle should be larger in future studies. The data should be confirmed from a multicentre perspective. The analysis should be performed also in non-PGT cycles and/or including patients who did not produce blastocysts, in order to investigate a putative association between women's reproductive history with outcomes other than euploidy and LBRs. WIDER IMPLICATIONS OF THE FINDINGS These data are critical to counsel infertile couples before, during and after a PGT-A cycle, especially to prevent treatment discontinuation due to previous adverse reproductive events. Beyond the 'maternal age effect', the causes of idiopathic recurrent pregnancy loss (RPL) and RIF are likely to be endometrial receptivity and selectivity issues; transferring euploid blastocysts might reduce the risk of a further miscarriage, but more information beyond euploidy are required to improve the prognosis in case of RIF. STUDY FUNDING/COMPETING INTEREST(S) No funding was received and there are no competing interests. TRIAL REGISTRATION NUMBER N/A.
Collapse
Affiliation(s)
| | | | - Lisa Dovere
- GeneraLife IVF, Clinica Valle Giulia, 00197 Rome, Italy
| | - Luisa Tacconi
- GeneraLife IVF, Clinica Valle Giulia, 00197 Rome, Italy
| | - Daria Soscia
- GeneraLife IVF, Clinica Valle Giulia, 00197 Rome, Italy
| | | | | | | | | | - Laura Rienzi
- GeneraLife IVF, Clinica Valle Giulia, 00197 Rome, Italy
| | | |
Collapse
|
26
|
Blyth U, Craciunas L, Hudson G, Choudhary M. Maternal germline factors associated with aneuploid pregnancy loss: a systematic review. Hum Reprod Update 2021; 27:866-884. [PMID: 33969392 DOI: 10.1093/humupd/dmab010] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Revised: 03/02/2021] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Miscarriage describes the spontaneous loss of pregnancy before the threshold of viability; the vast majority occur before 12 weeks of gestation. Miscarriage affects one in four couples and is the most common complication of pregnancy. Chromosomal abnormalities of the embryo are identified in ∼50% of first trimester miscarriages; aneuploidy accounts for 86% of these cases. The majority of trisomic miscarriages are of maternal origin with errors occurring during meiotic division of the oocytes. Chromosome segregation errors in oocytes may be sporadic events secondary to advancing maternal age; however, there is increasing evidence to suggest possible maternal germline contributions to this. OBJECTIVE AND RATIONALE The objective of this review was to appraise critically the existing evidence relating to maternal germline factors associated with pregnancy loss secondary to embryo aneuploidy, identify limitations in the current evidence base and establish areas requiring further research. SEARCH METHODS The initial literature search was performed in September 2019 and updated in January 2021 using the electronic databases OVID MEDLINE, EMBASE and the Cochrane Library. No time or language restrictions were applied to the searches and only primary research was included. Participants were women who had suffered pregnancy loss secondary to numerical chromosomal abnormalities of the embryo. Study identification and subsequent data extraction were performed by two authors independently. The Newcastle-Ottawa Scale was used to judge the quality of the included studies. The results were synthesized narratively. OUTCOMES The literature search identified 2198 titles once duplicates were removed, of which 21 were eligible for inclusion in this systematic review. They reported on maternal germline factors having variable degrees of association with pregnancy loss of aneuploid origin. The Online Mendelian Inheritance in Man (OMIM) gene ontology database was used as a reference to establish the functional role currently attributed to the genes reported. The majority of the cases reported and included were secondary to the inheritance of maternal structural factors such as Robertsonian translocations, deletions and insertions. Germline factors with a plausible role in aneuploid pregnancy loss of maternal origin included skewed X-inactivation and CGG repeats in the fragile X mental retardation (FMR1) gene. Studies that reported the association of single gene mutations with aneuploid pregnancy loss were conflicting. Single gene mutations with an uncertain or no role in aneuploid pregnancy loss included mutations in synaptonemal complex protein 3 (SYCP3), mitotic polo-like kinase 4 (PLK4) and meiotic stromal antigen 3 (STAG3) spindle integrity variants and 5,10-methylenetetrahydrofolate reductase (MTHFR). WIDER IMPLICATIONS Identifying maternal genetic factors associated with an increased risk of aneuploidy will expand our understanding of cell division, non-disjunction and miscarriage secondary to embryo aneuploidy. The candidate germline factors identified may be incorporated in a screening panel for women suffering miscarriage of aneuploidy aetiology to facilitate counselling for subsequent pregnancies.
Collapse
Affiliation(s)
- Ursula Blyth
- Newcastle Fertility Centre at Life, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, UK
| | - Laurentiu Craciunas
- Population Health Sciences Institute, Newcastle University, Newcastle upon Tyne, UK
| | - Gavin Hudson
- Biosciences Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK
| | - Meenakshi Choudhary
- Newcastle Fertility Centre at Life, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, UK
| |
Collapse
|
27
|
Shukla V, Høffding MK, Hoffmann ER. Genome diversity and instability in human germ cells and preimplantation embryos. Semin Cell Dev Biol 2021; 113:132-147. [PMID: 33500205 PMCID: PMC8097364 DOI: 10.1016/j.semcdb.2020.12.007] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Accepted: 12/18/2020] [Indexed: 12/26/2022]
Abstract
Genome diversity is essential for evolution and is of fundamental importance to human health. Generating genome diversity requires phases of DNA damage and repair that can cause genome instability. Humans have a high incidence of de novo congenital disorders compared to other organisms. Recent access to eggs, sperm and preimplantation embryos is revealing unprecedented rates of genome instability that may result in infertility and de novo mutations that cause genomic imbalance in at least 70% of conceptions. The error type and incidence of de novo mutations differ during developmental stages and are influenced by differences in male and female meiosis. In females, DNA repair is a critical factor that determines fertility and reproductive lifespan. In males, aberrant meiotic recombination causes infertility, embryonic failure and pregnancy loss. Evidence suggest germ cells are remarkably diverse in the type of genome instability that they display and the DNA damage responses they deploy. Additionally, the initial embryonic cell cycles are characterized by a high degree of genome instability that cause congenital disorders and may limit the use of CRISPR-Cas9 for heritable genome editing.
Collapse
Affiliation(s)
- Vallari Shukla
- DNRF Center for Chromosome Stability, Department of Cellular and Molecular Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Denmark
| | - Miya Kudo Høffding
- DNRF Center for Chromosome Stability, Department of Cellular and Molecular Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Denmark
| | - Eva R Hoffmann
- DNRF Center for Chromosome Stability, Department of Cellular and Molecular Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Denmark.
| |
Collapse
|
28
|
Tyc KM, El Yakoubi W, Bag A, Landis J, Zhan Y, Treff NR, Scott RT, Tao X, Schindler K, Xing J. Exome sequencing links CEP120 mutation to maternally derived aneuploid conception risk. Hum Reprod 2021; 35:2134-2148. [PMID: 32772081 DOI: 10.1093/humrep/deaa148] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2020] [Revised: 05/14/2020] [Indexed: 12/22/2022] Open
Abstract
STUDY QUESTION What are the genetic factors that increase the risk of aneuploid egg production? SUMMARY ANSWER A non-synonymous variant rs2303720 within centrosomal protein 120 (CEP120) disrupts female meiosis in vitro in mouse. WHAT IS KNOWN ALREADY The production of aneuploid eggs, with an advanced maternal age as an established contributing factor, is the major cause of IVF failure, early miscarriage and developmental anomalies. The identity of maternal genetic variants contributing to egg aneuploidy irrespective of age is missing. STUDY DESIGN, SIZE, DURATION Patients undergoing fertility treatment (n = 166) were deidentified and selected for whole-exome sequencing. PARTICIPANTS/MATERIALS, SETTING, METHODS Patients self-identified their ethnic groups and their ages ranged from 22 to 49 years old. The study was performed using genomes from White, non-Hispanic patients divided into controls (97) and cases (69) according to the number of aneuploid blastocysts derived during each IVF procedure. Following a gene prioritization strategy, a mouse oocyte system was used to validate the functional significance of the discovered associated genetic variants. MAIN RESULTS AND THE ROLE OF CHANCE Patients producing a high proportion of aneuploid blastocysts (considered aneuploid if they missed any of the 40 chromatids or had extra copies) were found to carry a higher mutational burden in genes functioning in cytoskeleton and microtubule pathways. Validation of the functional significance of a non-synonymous variant rs2303720 within Cep120 on mouse oocyte meiotic maturation revealed that ectopic expression of CEP120:p.Arg947His caused decreased spindle microtubule nucleation efficiency and increased incidence of aneuploidy. LIMITATIONS, REASONS FOR CAUTION Functional validation was performed using the mouse oocyte system. Because spindle building pathways differ between mouse and human oocytes, the defects we observed upon ectopic expression of the Cep120 variant may alter mouse oocyte meiosis differently than human oocyte meiosis. Further studies using knock-in 'humanized' mouse models and in human oocytes will be needed to translate our findings to human system. Possible functional differences of the variant between ethnic groups also need to be investigated. WIDER IMPLICATIONS OF THE FINDINGS Variants in centrosomal genes appear to be important contributors to the risk of maternal aneuploidy. Functional validation of these variants will eventually allow prescreening to select patients that have better chances to benefit from preimplantation genetic testing. STUDY FUNDING/COMPETING INTEREST(S) This study was funded through R01-HD091331 to K.S. and J.X. and EMD Serono Grant for Fertility Innovation to N.R.T. N.R.T. is a shareholder and an employee of Genomic Prediction. TRIAL REGISTRATION NUMBER N/A.
Collapse
Affiliation(s)
- Katarzyna M Tyc
- Department of Genetics, Rutgers, The State University of New Jersey, Piscataway, NJ 08854, USA.,Human Genetics Institute of New Jersey, Rutgers, The State University of New Jersey, Piscataway, NJ 08854, USA
| | - Warif El Yakoubi
- Department of Genetics, Rutgers, The State University of New Jersey, Piscataway, NJ 08854, USA.,Human Genetics Institute of New Jersey, Rutgers, The State University of New Jersey, Piscataway, NJ 08854, USA
| | - Aishee Bag
- Department of Genetics, Rutgers, The State University of New Jersey, Piscataway, NJ 08854, USA
| | - Jessica Landis
- Foundation for Embryonic Competence, Basking Ridge, NJ 07920, USA
| | - Yiping Zhan
- Foundation for Embryonic Competence, Basking Ridge, NJ 07920, USA
| | - Nathan R Treff
- Reproductive Medicine Associates of New Jersey, Basking Ridge, NJ 07920, USA
| | - Richard T Scott
- Reproductive Medicine Associates of New Jersey, Basking Ridge, NJ 07920, USA
| | - Xin Tao
- Foundation for Embryonic Competence, Basking Ridge, NJ 07920, USA
| | - Karen Schindler
- Department of Genetics, Rutgers, The State University of New Jersey, Piscataway, NJ 08854, USA.,Human Genetics Institute of New Jersey, Rutgers, The State University of New Jersey, Piscataway, NJ 08854, USA
| | - Jinchuan Xing
- Department of Genetics, Rutgers, The State University of New Jersey, Piscataway, NJ 08854, USA.,Human Genetics Institute of New Jersey, Rutgers, The State University of New Jersey, Piscataway, NJ 08854, USA
| |
Collapse
|
29
|
Paonessa M, Borini A, Coticchio G. Genetic causes of preimplantation embryo developmental failure. Mol Reprod Dev 2021; 88:338-348. [PMID: 33843124 DOI: 10.1002/mrd.23471] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Revised: 02/27/2021] [Accepted: 03/23/2021] [Indexed: 12/13/2022]
Abstract
Embryo development requires orchestrated events, finely regulated at the molecular and cellular level by mechanisms which are progressively emerging from animal studies. With progress in genetic technologies-such as genome editing and single-cell RNA analysis-we can now assess embryo gene expression with increased precision and gain new insights into complex processes until recently difficult to explore. Multiple genes and regulative pathways have been identified for each developmental stage. We have learned that embryos with undisturbed and timely gene expression have higher chances of successful development. For example, selected genes are highly expressed during the first stages, being involved in cell adhesion, cell cycle, and regulation of transcription; other genes are instead crucial for lineage specification and therefore expressed at later stages. Due to ethical constraints, studies on human embryos remain scarce, mainly descriptive, and unable to provide functional evidence. This highlights the importance of animal studies as basic knowledge to test and appraise in a clinical context. In this review, we report on preimplantation development with a focus on genes whose impairment leads to developmental arrest. Preconceptional genetic screening could identify loss-of-function mutations of these genes; thereby, novel biomarkers of embryo quality could be adopted to improve diagnosis and treatment of infertility.
Collapse
Affiliation(s)
- Mariagrazia Paonessa
- 9.Baby, Family and Fertility Center, Bologna, Italy.,Casa di Cura Candela Spa, Palermo, Italy
| | | | | |
Collapse
|
30
|
Coticchio G, Pennetta F, Rizzo R, Tarozzi N, Nadalini M, Orlando G, Centonze C, Gioacchini G, Borini A. Embryo morphokinetic score is associated with biomarkers of developmental competence and implantation. J Assist Reprod Genet 2021; 38:1737-1743. [PMID: 33821429 DOI: 10.1007/s10815-021-02162-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Accepted: 03/18/2021] [Indexed: 12/01/2022] Open
Abstract
PURPOSE To study embryo morphokinetics in relation to release in spent media of molecules with possible roles in development and implantation (miR-20a, miR-30c, and sHLA-G). METHODS Data were obtained from embryos generated in standard IVF and ICSI cycles. The Eeva system was used for embryo assessment, based on early morphokinetic parameters and producing a score (1-5, best-worst) corresponding to higher/medium/lower chances of development to blastocyst. miRNAs - mm miR-20a-5p and miR-30c-5p - and sHLA-G were quantified in 25 μl of spent blastocyst media (SBM) collected before vitrification or transfer. Statistical analyses were performed applying Kolmogorov-Smirnov, Shapiro-Wilk, and Spearman's correlation coefficient tests, where appropriate. RESULTS SBM were collected from a total of 172 viable blastocysts. Their analysis showed that concentration of miR-20a was progressively lower as Eeva score increased and probability of development to blastocyst decreased (P = 0.016). The opposite trend was observed in the case of miR-30c, i.e., concentration was higher as score increased and chances of development to blastocyst decreased (P = 0.004). Analysis of sHLA-G revealed a negative correlation with Eeva score, i.e., levels were progressively lower as Eeva score increased and probability of development to blastocyst decreased (R = - 0.388, N = 141, P = 0.001). CONCLUSION Our data suggest that morphokinetic algorithms that predict development to blastocyst stage, in fact, also identify embryos with molecular and cellular profiles more consistent with developmental functions.
Collapse
Affiliation(s)
- Giovanni Coticchio
- 9.baby Family and Fertility Center, Via Dante, 15, 40125, Bologna, Italy.
| | - Francesca Pennetta
- 9.baby Family and Fertility Center, Via Dante, 15, 40125, Bologna, Italy
- Simple Departmental Operative Unit, Reproductive Pathophysiology, Anastasia Guerriero Hospital, Marcianise, Caserta, Italy
| | - Roberta Rizzo
- Section of Microbiology and Medical Genetics, Department of Medical Sciences, University of Ferrara, Ferrara, Italy
| | - Nicoletta Tarozzi
- 9.baby Family and Fertility Center, Via Dante, 15, 40125, Bologna, Italy
| | - Marco Nadalini
- 9.baby Family and Fertility Center, Via Dante, 15, 40125, Bologna, Italy
| | | | | | - Giorgia Gioacchini
- Department of Life and Environmental Sciences, Polytechnic University of Marche, Ancona, Italy
| | - Andrea Borini
- 9.baby Family and Fertility Center, Via Dante, 15, 40125, Bologna, Italy
| |
Collapse
|
31
|
Wartosch L, Schindler K, Schuh M, Gruhn JR, Hoffmann ER, McCoy RC, Xing J. Origins and mechanisms leading to aneuploidy in human eggs. Prenat Diagn 2021; 41:620-630. [PMID: 33860956 PMCID: PMC8237340 DOI: 10.1002/pd.5927] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Revised: 01/02/2021] [Accepted: 02/21/2021] [Indexed: 11/18/2022]
Abstract
The gain or loss of a chromosome-or aneuploidy-acts as one of the major triggers for infertility and pregnancy loss in humans. These chromosomal abnormalities affect more than 40% of eggs in women at both ends of the age spectrum, that is, young girls as well as women of advancing maternal age. Recent studies in human oocytes and embryos using genomics, cytogenetics, and in silico modeling all provide new insight into the rates and potential genetic and cellular factors associated with aneuploidy at varying stages of development. Here, we review recent studies that are shedding light on potential molecular mechanisms of chromosome missegregation in oocytes and embryos across the entire female reproductive life span.
Collapse
Affiliation(s)
- Lena Wartosch
- Department of MeiosisMax Planck Institute for Biophysical ChemistryGöttingenGermany
| | - Karen Schindler
- Department of GeneticsRutgers, The State University of New JerseyPiscatawayNew JerseyUSA
- Human Genetics Institute of New JerseyRutgers, The State University of New JerseyPiscatawayNew JerseyUSA
| | - Melina Schuh
- Department of MeiosisMax Planck Institute for Biophysical ChemistryGöttingenGermany
| | - Jennifer R. Gruhn
- DNRF Center for Chromosome StabilityDepartment of Cellular and Molecular MedicineFaculty of Health and Medical SciencesUniversity of CopenhagenDenmark
| | - Eva R. Hoffmann
- DNRF Center for Chromosome StabilityDepartment of Cellular and Molecular MedicineFaculty of Health and Medical SciencesUniversity of CopenhagenDenmark
| | - Rajiv C. McCoy
- Department of BiologyJohns Hopkins UniversityBaltimoreMarylandUSA
| | - Jinchuan Xing
- Department of GeneticsRutgers, The State University of New JerseyPiscatawayNew JerseyUSA
- Human Genetics Institute of New JerseyRutgers, The State University of New JerseyPiscatawayNew JerseyUSA
| |
Collapse
|
32
|
Levy B, Hoffmann ER, McCoy RC, Grati FR. Chromosomal mosaicism: Origins and clinical implications in preimplantation and prenatal diagnosis. Prenat Diagn 2021; 41:631-641. [PMID: 33720449 DOI: 10.1002/pd.5931] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Revised: 03/03/2021] [Accepted: 03/06/2021] [Indexed: 12/18/2022]
Abstract
The diagnosis of chromosomal mosaicism in the preimplantation and prenatal stage is fraught with uncertainty and multiple factors need to be considered in order to gauge the likely impact. The clinical effects of chromosomal mosaicism are directly linked to the type of the imbalance (size, gene content, and copy number), the timing of the initial event leading to mosaicism during embryogenesis/fetal development, the distribution of the abnormal cells throughout the various tissues within the body as well as the ratio of normal/abnormal cells within each of those tissues. Additional factors such as assay noise and culture artifacts also have an impact on the significance and management of mosaic cases. Genetic counseling is an important part of educating patients about the likelihood of having a liveborn with a chromosome abnormality and these risks differ according to the time of ascertainment and the tissue where the mosaic cells were initially discovered. Each situation needs to be assessed on a case-by-case basis and counseled accordingly. This review will discuss the clinical impact of finding mosaicism through: embryo biopsy, chorionic villus sampling, amniocentesis, and noninvasive prenatal testing using cell-free DNA.
Collapse
Affiliation(s)
- Brynn Levy
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, New York, USA
| | - Eva R Hoffmann
- Department of Cellular and Molecular Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Rajiv C McCoy
- Department of Biology, Johns Hopkins University, Baltimore, Maryland, USA
| | - Francesca R Grati
- Research and Development, Cytogenetics and Medical Genetics Unit, TOMA Advanced Biomedical Assays, S.p.A. (Impact Lab), Busto Arsizio, Varese, Italy
| |
Collapse
|
33
|
van Marion ES, Speksnijder JP, Hoek J, Boellaard WPA, Dinkelman-Smit M, Chavli EA, Steegers-Theunissen RPM, Laven JSE, Baart EB. Time-lapse imaging of human embryos fertilized with testicular sperm reveals an impact on the first embryonic cell cycle. Biol Reprod 2021; 104:1218-1227. [PMID: 33690817 PMCID: PMC8181962 DOI: 10.1093/biolre/ioab031] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2020] [Revised: 02/08/2021] [Accepted: 02/18/2021] [Indexed: 11/14/2022] Open
Abstract
Testicular sperm is increasingly used during in vitro fertilization treatment. Testicular sperm has the ability to fertilize the oocyte after intracytoplasmic sperm injection (ICSI), but they have not undergone maturation during epididymal transport. Testicular sperm differs from ejaculated sperm in terms of chromatin maturity, incidence of DNA damage, and RNA content. It is not fully understood what the biological impact is of using testicular sperm, on fertilization, preimplantation embryo development, and postimplantation development. Our goal was to investigate differences in human preimplantation embryo development after ICSI using testicular sperm (TESE-ICSI) and ejaculated sperm. We used time-lapse embryo culture to study these possible differences. Embryos (n = 639) originating from 208 couples undergoing TESE-ICSI treatment were studied and compared to embryos (n = 866) originating from 243 couples undergoing ICSI treatment with ejaculated sperm. Using statistical analysis with linear mixed models, we observed that pronuclei appeared 0.55 h earlier in TESE-ICSI embryos, after which the pronuclear stage lasted 0.55 h longer. Also, significantly more TESE-ICSI embryos showed direct unequal cleavage from the 1-cell stage to the 3-cell stage. TESE-ICSI embryos proceeded faster through the cleavage divisions to the 5- and the 6-cell stage, but this effect disappeared when we adjusted our model for maternal factors. In conclusion, sperm origin affects embryo development during the first embryonic cell cycle, but not developmental kinetics to the 8-cell stage. Our results provide insight into the biological differences between testicular and ejaculated sperm and their impact during human fertilization.
Collapse
Affiliation(s)
- E S van Marion
- Division of Reproductive Endocrinology and Infertility, Department of Obstetrics and Gynaecology, Erasmus MC, University Medical Center, Rotterdam, The Netherlands
| | - J P Speksnijder
- Division of Reproductive Endocrinology and Infertility, Department of Obstetrics and Gynaecology, Erasmus MC, University Medical Center, Rotterdam, The Netherlands
| | - J Hoek
- Department of Obstetrics and Gynaecology, Erasmus MC, University Medical Center, Rotterdam, The Netherlands
| | - W P A Boellaard
- Department of Urology, Erasmus MC, University Medical Center, Rotterdam, The Netherlands
| | - M Dinkelman-Smit
- Department of Urology, Erasmus MC, University Medical Center, Rotterdam, The Netherlands
| | - E A Chavli
- Division of Reproductive Endocrinology and Infertility, Department of Obstetrics and Gynaecology, Erasmus MC, University Medical Center, Rotterdam, The Netherlands
| | - R P M Steegers-Theunissen
- Department of Obstetrics and Gynaecology, Erasmus MC, University Medical Center, Rotterdam, The Netherlands
| | - J S E Laven
- Division of Reproductive Endocrinology and Infertility, Department of Obstetrics and Gynaecology, Erasmus MC, University Medical Center, Rotterdam, The Netherlands
| | - E B Baart
- Division of Reproductive Endocrinology and Infertility, Department of Obstetrics and Gynaecology, Erasmus MC, University Medical Center, Rotterdam, The Netherlands.,Department of Developmental Biology, Erasmus MC, University Medical Center, Rotterdam, The Netherlands
| |
Collapse
|
34
|
Ravisankar S, Ting AY, Murphy MJ, Redmayne N, Wang D, McArthur CA, Takahashi DL, Kievit P, Chavez SL, Hennebold JD. Short-term Western-style diet negatively impacts reproductive outcomes in primates. JCI Insight 2021; 6:138312. [PMID: 33616080 PMCID: PMC7934943 DOI: 10.1172/jci.insight.138312] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Accepted: 01/13/2021] [Indexed: 12/19/2022] Open
Abstract
A maternal Western-style diet (WSD) is associated with poor reproductive outcomes, but whether this is from the diet itself or underlying metabolic dysfunction is unknown. Here, we performed a longitudinal study using regularly cycling female rhesus macaques (n = 10) that underwent 2 consecutive in vitro fertilization (IVF) cycles, one while consuming a low-fat diet and another 6–8 months after consuming a high-fat WSD. Metabolic data were collected from the females prior to each IVF cycle. Follicular fluid (FF) and oocytes were assessed for cytokine/steroid levels and IVF potential, respectively. Although transition to a WSD led to weight gain and increased body fat, no difference in insulin levels was observed. A significant decrease in IL-1RA concentration and the ratio of cortisol/cortisone was detected in FF after WSD intake. Despite an increased probability of isolating mature oocytes, a 44% reduction in blastocyst number was observed with WSD consumption, and time-lapse imaging revealed delayed mitotic timing and multipolar divisions. RNA sequencing of blastocysts demonstrated dysregulation of genes involved in RNA binding, protein channel activity, mitochondrial function and pluripotency versus cell differentiation after WSD consumption. Thus, short-term WSD consumption promotes a proinflammatory intrafollicular microenvironment that is associated with impaired preimplantation development in the absence of large-scale metabolic changes.
Collapse
Affiliation(s)
- Sweta Ravisankar
- Department of Cell, Developmental & Cancer Biology, Graduate Program in Molecular & Cellular Biosciences, Oregon Health & Science University School of Medicine, Portland, Oregon, USA.,Division of Reproductive & Developmental Sciences, Oregon National Primate Research Center, Beaverton, Oregon, USA
| | - Alison Y Ting
- Division of Reproductive & Developmental Sciences, Oregon National Primate Research Center, Beaverton, Oregon, USA.,21st Century Medicine Inc., Fontana, California, USA
| | - Melinda J Murphy
- Division of Reproductive & Developmental Sciences, Oregon National Primate Research Center, Beaverton, Oregon, USA
| | - Nash Redmayne
- Division of Reproductive & Developmental Sciences, Oregon National Primate Research Center, Beaverton, Oregon, USA
| | - Dorothy Wang
- Division of Reproductive & Developmental Sciences, Oregon National Primate Research Center, Beaverton, Oregon, USA
| | - Carrie A McArthur
- Division of Cardiometabolic Health, Oregon National Primate Research Center, Beaverton, Oregon, USA
| | - Diana L Takahashi
- Division of Cardiometabolic Health, Oregon National Primate Research Center, Beaverton, Oregon, USA
| | - Paul Kievit
- Division of Cardiometabolic Health, Oregon National Primate Research Center, Beaverton, Oregon, USA
| | - Shawn L Chavez
- Division of Reproductive & Developmental Sciences, Oregon National Primate Research Center, Beaverton, Oregon, USA.,Department of Molecular & Medical Genetics, Oregon Health & Science University School of Medicine, Portland, Oregon, USA.,Department of Obstetrics & Gynecology, Oregon Health & Science University School of Medicine, Portland, Oregon, USA
| | - Jon D Hennebold
- Division of Reproductive & Developmental Sciences, Oregon National Primate Research Center, Beaverton, Oregon, USA.,Department of Obstetrics & Gynecology, Oregon Health & Science University School of Medicine, Portland, Oregon, USA
| |
Collapse
|
35
|
Cimadomo D, Capalbo A, Scarica C, Sosa Fernandez L, Rienzi L, Ciriminna R, Minasi MG, Novelli A, De Santis L, Zuccarello D. When embryology meets genetics: the definition of developmentally incompetent preimplantation embryos (DIPE)-the consensus of two Italian scientific societies. J Assist Reprod Genet 2021; 38:319-331. [PMID: 33236289 PMCID: PMC7884494 DOI: 10.1007/s10815-020-02015-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Accepted: 11/12/2020] [Indexed: 02/06/2023] Open
Abstract
A clear definition of developmentally incompetent preimplantation embryo (DIPE) in literature is still missing, while several scientific societies are discussing this challenging topic. From both a clinical and scientific perspective, the identification of embryos unfit for reproductive purpose is crucial. This aim should be pursued in light of all diagnostic technologies for embryo evaluation, encompassing also genetic analyses, of recent implementation in IVF. The Italian context is characterized by an unusual scenario: embryos can be discarded only if not viable and cannot be used for research purposes either. Therefore, thousands of embryos, diagnosed as affected and/or aneuploid as resulting from preimplantation genetic testing (PGT) and clinically not utilizable, are cryopreserved and stored indefinitely, with important psychological, legal, and financial implications. With the aim of updating the definition of DIPE, also on the basis of the embryo genetic status, the Italian Society of Embryology, Reproduction and Research (SIERR) and the Italian Society of Human Genetic (SIGU) reviewed the literature on this topic, found a consensus, and produced a list of relevant criteria.
Collapse
Affiliation(s)
- Danilo Cimadomo
- GeneraLife IVF centers, Clinica Valle Giulia, via G. de Notaris 2b, 00197, Rome, Italy.
| | | | - Catello Scarica
- Center for Reproductive Medicine, Casa di Cura Villa Salaria in Partnership with Institut Marques, Rome, Italy
| | | | - Laura Rienzi
- GeneraLife IVF centers, Clinica Valle Giulia, via G. de Notaris 2b, 00197, Rome, Italy
| | | | | | - Antonio Novelli
- Laboratory of Medical Genetics, IRCCS-Bambino Gesù Children's Hospital, Rome, Italy
| | - Lucia De Santis
- Department of Obstetrics & Gynecology, IVF Unit, San Raffaele Scientific Institute, Vita-Salute San Raffaele University, Milan, Italy
| | | |
Collapse
|
36
|
Masset H, Tšuiko O, Vermeesch JR. Genome-wide abnormalities in embryos: Origins and clinical consequences. Prenat Diagn 2021; 41:554-563. [PMID: 33524193 DOI: 10.1002/pd.5895] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Revised: 12/03/2020] [Accepted: 12/30/2020] [Indexed: 12/25/2022]
Abstract
Ploidy or genome-wide chromosomal anomalies such as triploidy, diploid/triploid mixoploidy, chimerism, and genome-wide uniparental disomy are the cause of molar pregnancies, embryonic lethality, and developmental disorders. While triploidy and genome-wide uniparental disomy can be ascribed to fertilization or meiotic errors, the mechanisms causing mixoploidy and chimerism remain shrouded in mystery. Different models have been proposed, but all remain hypothetical and controversial, are deduced from the developmental persistent genomic constitutions present in the sample studied and lack direct evidence. New single-cell genomic methodologies, such as single-cell genome-wide haplotyping, provide an extended view of the constitution of normal and abnormal embryos and have further pinpointed the existence of mixoploidy in cleavage-stage embryos. Based on those recent findings, we suggest that genome-wide anomalies, which persist in fetuses and patients, can for a large majority be explained by a noncanonical first zygotic cleavage event, during which maternal and paternal genomes in a single zygote, segregate to different blastomeres. This process, termed heterogoneic division, provides an overarching theoretical basis for the different presentations of mixoploidy and chimerism.
Collapse
Affiliation(s)
- Heleen Masset
- Department of Human Genetics, Laboratory for Cytogenetics and Genome Research, KU Leuven, Leuven, Belgium
| | - Olga Tšuiko
- Department of Human Genetics, Laboratory for Cytogenetics and Genome Research, KU Leuven, Leuven, Belgium
| | - Joris R Vermeesch
- Department of Human Genetics, Laboratory for Cytogenetics and Genome Research, KU Leuven, Leuven, Belgium.,Center of Human Genetics, University Hospitals of Leuven, Leuven, Belgium
| |
Collapse
|
37
|
Time of morulation and trophectoderm quality are predictors of a live birth after euploid blastocyst transfer: a multicenter study. Fertil Steril 2020; 112:1080-1093.e1. [PMID: 31843084 DOI: 10.1016/j.fertnstert.2019.07.1322] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2019] [Revised: 07/11/2019] [Accepted: 07/22/2019] [Indexed: 12/15/2022]
Abstract
OBJECTIVE To investigate whether the morphodynamic characterization of a euploid blastocyst's development allows a higher prediction of a live birth after single-embryo-transfer (SET). DESIGN Observational cohort study conducted in two phases: training and validation. SETTING Private in vitro fertilization centers. PATIENT(S) Euploid blastocysts: 511 and 319 first vitrified-warmed SETs from 868 and 546 patients undergoing preimplantation genetic testing for aneuploidies (PGT-A) in the training and validation phase, respectively. INTERVENTION(S) Data collected from time of polar body extrusion to time of starting blastulation, and trophectoderm and inner-cell-mass static morphology in all embryos cultured in a specific time-lapse incubator with a continuous medium. Logistic regressions conducted to outline the variables showing a statistically significant association with live birth. In the validation phase, these variables were tested in an independent data set. MAIN OUTCOME MEASURE(S) Live births per SET. RESULT(S) The average live birth rate (LBR) in the training set was 40% (N = 207/511). Only time of morulation (tM) and trophectoderm quality were outlined as putative predictors of live birth at two IVF centers. In the validation set, the euploid blastocysts characterized by tM <80 hours and high-quality trophectoderm resulted in a LBR of 55.2% (n = 37/67), while those with tM ≥ 80 hours and a low-quality trophectoderm resulted in a LBR of 25.5% (N = 13/51). CONCLUSION(S) Time of morulation and trophectoderm quality are better predictors of a euploid blastocyst's reproductive competence. Our evidence was reproducible across different centers under specific culture conditions. These data support the crucial role of morulation for embryo development, a stage that involves massive morphologic, cellular, and molecular changes and deserves more investigation.
Collapse
|
38
|
Wang F, Liu Y. Identification of key genes, regulatory factors, and drug target genes of recurrent implantation failure (RIF). Gynecol Endocrinol 2020; 36:448-455. [PMID: 31646911 DOI: 10.1080/09513590.2019.1680622] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Objective: Recurrent implantation failure (RIF) exacerbates the physical trauma of infertile women that undergone in vitro fertilization-embryo transfer (IVF-ET). We aimed to identify the key genes, regulatory factors, and drug target genes involved in the RIF.Methods: The dataset GSE58144 that obtained from the Gene Expression Omnibus mainly contained 43 RIF and 72 control endometrial samples. Differently expressed genes (DEGs) between RIF and control groups were firstly analyzed, followed by the pathway and Gene Ontology (GO) enrichment analysis. Then, protein-protein interaction (PPI) network and miRNA-transcript factor (TF)-DEGs network were established. Finally, a drug-target interaction network was constructed.Results: A total of 399 DEGs were identified between the RIF and controls. In the PPI and key module network, UBE2I, PLK4, XPO1, AURKB, and NUP107 were identified as the hub genes, which mainly enriched in RNA transport and cell division cycle-related pathways and GO items. In the miRNA-TF-DEGs network, E2F4, SIN3A, miRNA489, miRNA199A, miRNA369-3P, miRNA422, and miRNA522 were considered as the key regulatory factors during RIF. In addition, HTR1A, NR3C1, and GABRA3 were the main targets of the drugs annotated in DrugBank.Conclusion: The effects of PLK4, XPO1, AURKB, and NUP107 on the RIF may be via affecting the proliferation and differentiation of endometrial stromal cells. Besides, SIN3A and miRNA199A may be crucial for embryo implantation. In addition, NR3C1 may be used as a possible target for the clinical therapy of RIF.
Collapse
Affiliation(s)
- Fang Wang
- Department of Gynaecology and Obstetrics, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Yaofang Liu
- Department of Gynaecology and Obstetrics, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| |
Collapse
|
39
|
Albertini DF. Novel insights into the genetics of early human development: PGT as a catalyst for reform. J Assist Reprod Genet 2020; 37:495-496. [PMID: 32219599 PMCID: PMC7100432 DOI: 10.1007/s10815-020-01759-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
|
40
|
Alternative patterns of partial embryo compaction: prevalence, morphokinetic history and possible implications. Reprod Biomed Online 2020; 40:347-354. [DOI: 10.1016/j.rbmo.2019.11.011] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2019] [Revised: 11/18/2019] [Accepted: 11/21/2019] [Indexed: 12/11/2022]
|
41
|
Destouni A, Dimitriadou E, Masset H, Debrock S, Melotte C, Van Den Bogaert K, Zamani Esteki M, Ding J, Voet T, Denayer E, de Ravel T, Legius E, Meuleman C, Peeraer K, Vermeesch JR. Genome-wide haplotyping embryos developing from 0PN and 1PN zygotes increases transferrable embryos in PGT-M. Hum Reprod 2019; 33:2302-2311. [PMID: 30383227 PMCID: PMC6238370 DOI: 10.1093/humrep/dey325] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2018] [Accepted: 10/14/2018] [Indexed: 02/06/2023] Open
Abstract
STUDY QUESTION Can genome-wide haplotyping increase success following preimplantation genetic testing for a monogenic disorder (PGT-M) by including zygotes with absence of pronuclei (0PN) or the presence of only one pronucleus (1PN)? SUMMARY ANSWER Genome-wide haplotyping 0PNs and 1PNs increases the number of PGT-M cycles reaching embryo transfer (ET) by 81% and the live-birth rate by 75%. WHAT IS KNOWN ALREADY Although a significant subset of 0PN and 1PN zygotes can develop into balanced, diploid and developmentally competent embryos, they are usually discarded because parental diploidy detection is not part of the routine work-up of PGT-M. STUDY DESIGN, SIZE, DURATION This prospective cohort study evaluated the pronuclear number in 2229 zygotes from 2337 injected metaphase II (MII) oocytes in 268 cycles. PGT-M for 0PN and 1PN embryos developing into Day 5/6 blastocysts with adequate quality for vitrification was performed in 42 of the 268 cycles (15.7%). In these 42 cycles, we genome-wide haplotyped 216 good quality embryos corresponding to 49 0PNs, 15 1PNs and 152 2PNs. The reported outcomes include parental contribution to embryonic ploidy, embryonic aneuploidy, genetic diagnosis for the monogenic disorder, cycles reaching ETs, pregnancy and live birth rates (LBR) for unaffected offspring. PARTICIPANTS/MATERIALS, SETTING, METHODS Blastomere DNA was whole-genome amplified and hybridized on the Illumina Human CytoSNP12V2.1.1 BeadChip arrays. Subsequently, genome-wide haplotyping and copy-number profiling was applied to investigate the embryonic genome architecture. Bi-parental, unaffected embryos were transferred regardless of their initial zygotic PN score. MAIN RESULTS AND THE ROLE OF CHANCE A staggering 75.51% of 0PN and 42.86% of 1PN blastocysts are diploid bi-parental allowing accurate genetic diagnosis for the monogenic disorder. In total, 31% (13/42) of the PGT-M cycles reached ET or could repeat ET with an unaffected 0PN or 1PN embryo. The LBR per initiated cycle increased from 9.52 to 16.67%. LIMITATIONS, REASONS FOR CAUTION The clinical efficacy of the routine inclusion of 0PN and 1PN zygotes in PGT-M cycles should be confirmed in larger cohorts from multicenter studies. WIDER IMPLICATIONS OF THE FINDINGS Genome-wide haplotyping allows the inclusion of 0PN and 1PN embryos and subsequently increases the cycles reaching ET following PGT-M and potentially PGT for aneuploidy (PGT-A) and chromosomal structural rearrangements (PGT-SR). Establishing measures of clinical efficacy could lead to an update of the ESHRE guidelines which advise against the use of these zygotes. STUDY FUNDING/COMPETING INTEREST(S) SymBioSys (PFV/10/016 and C1/018 to J.R.V. and T.V.), the Horizon 2020 WIDENLIFE: 692065 to J.R.V., T.V., E.D., A.D. and M.Z.E. M.Z.E., T.V. and J.R.V. co-invented haplarithmisis (‘Haplotyping and copy-number typing using polymorphic variant allelic frequencies’), which has been licensed to Agilent Technologies. H.M. is fully supported by the (FWO) (ZKD1543-ASP/16). The authors have no competing interests to declare.
Collapse
Affiliation(s)
- Aspasia Destouni
- Laboratory for Cytogenetics and Genome Research, Center for Human Genetics, University of Leuven, O&N I Herestraat 49, KU Leuven, Leuven, Belgium
| | - Eftychia Dimitriadou
- Department of Human Genetics, Center for Human Genetics, University Hospitals Leuven, O&N I Herestraat 49, KU Leuven, Leuven, Belgium
| | - Heleen Masset
- Laboratory for Cytogenetics and Genome Research, Center for Human Genetics, University of Leuven, O&N I Herestraat 49, KU Leuven, Leuven, Belgium
| | - Sophie Debrock
- University Hospitals Leuven, Leuven University Fertility Center, Herestraat 49, Leuven, Belgium
| | - Cindy Melotte
- Department of Human Genetics, Center for Human Genetics, University Hospitals Leuven, O&N I Herestraat 49, KU Leuven, Leuven, Belgium
| | - Kris Van Den Bogaert
- Department of Human Genetics, Center for Human Genetics, University Hospitals Leuven, O&N I Herestraat 49, KU Leuven, Leuven, Belgium
| | - Masoud Zamani Esteki
- Department of Human Genetics, Center for Human Genetics, University Hospitals Leuven, O&N I Herestraat 49, KU Leuven, Leuven, Belgium.,Maastricht University Medical Center, Department of Clinical Genetics, GROW School for Oncology and Developmental Biology, Maastricht, The Netherlands
| | - Jia Ding
- Laboratory for Cytogenetics and Genome Research, Center for Human Genetics, University of Leuven, O&N I Herestraat 49, KU Leuven, Leuven, Belgium
| | - Thiery Voet
- Laboratory of Reproductive Genomics, Center for Human Genetics, University of Leuven, O&N I Herestraat 49, KU Leuven, Leuven, Belgium.,Wellcome Sanger Institute, Wellcome Genome Campus Hinxton, Cambridgeshire, UK
| | - Ellen Denayer
- Department of Human Genetics, Center for Human Genetics, University Hospitals Leuven, O&N I Herestraat 49, KU Leuven, Leuven, Belgium
| | - Thomy de Ravel
- Department of Human Genetics, Center for Human Genetics, University Hospitals Leuven, O&N I Herestraat 49, KU Leuven, Leuven, Belgium
| | - Eric Legius
- Department of Human Genetics, Center for Human Genetics, University Hospitals Leuven, O&N I Herestraat 49, KU Leuven, Leuven, Belgium
| | - Christel Meuleman
- University Hospitals Leuven, Leuven University Fertility Center, Herestraat 49, Leuven, Belgium
| | - Karen Peeraer
- University Hospitals Leuven, Leuven University Fertility Center, Herestraat 49, Leuven, Belgium
| | - Joris R Vermeesch
- Laboratory for Cytogenetics and Genome Research, Center for Human Genetics, University of Leuven, O&N I Herestraat 49, KU Leuven, Leuven, Belgium.,Department of Human Genetics, Center for Human Genetics, University Hospitals Leuven, O&N I Herestraat 49, KU Leuven, Leuven, Belgium
| |
Collapse
|
42
|
Spindle and chromosome configuration analysis of human biopsied versus non-biopsied embryos by confocal laser scanning microscopy following vitrification. ZYGOTE 2019; 27:153-159. [PMID: 31060631 DOI: 10.1017/s0967199419000182] [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] [Indexed: 11/06/2022]
Abstract
SummaryThe aim of this study was to investigate the effects of zona drilling and biopsy on day 3 followed by vitrification on day 5 on the cytoskeleton and development of human embryos, by analysing survival rates and spindle and chromosome configurations by fluorescence and confocal laser scanning microscopy in human biopsied and non-biopsied embryos. In total, 98 human blastocysts (50 non-biopsied and 48 following biopsy on day 3) were vitrified on day 5 using either a commercial dimethyl sulphoxide (DMSO)-free vitrification kit or increasing concentrations of DMSO/EG (5%/5-10%/10-20%/20%). Following warming, the blastocysts were allowed to recover in culture for 24 h and were immunostained with α-tubulin, acetylated tubulin, and/or γ-tubulin antibodies in combination with 4',6-diamidino-2-phenylindole (DAPI). Labelled embryos were examined by both fluorescence and confocal laser scanning microscopy. The survival rates following warming (92% non-biopsied vs 83.3% biopsied) and the incidence of normal spindle chromosome configurations was not statistically different between the two groups (65.2% non-biopsied vs 59.2% biopsied, P>0.05). The incidence of spindle abnormalities including multipolarity, chromosome lagging, congression failure and chromosome bridging were also similar between the two groups (P>0.05). This study is the first to compare the incidence of cytoskeletal abnormalities in biopsied and non-biopsied human embryos following vitrification. We conclude that there was no significant difference in the survival rates and the incidence of spindle abnormalities between the two groups.
Collapse
|
43
|
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: 20] [Impact Index Per Article: 4.0] [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
|
44
|
Scarica C, Cimadomo D, Dovere L, Giancani A, Stoppa M, Capalbo A, Ubaldi FM, Rienzi L, Canipari R. An integrated investigation of oocyte developmental competence: expression of key genes in human cumulus cells, morphokinetics of early divisions, blastulation, and euploidy. J Assist Reprod Genet 2019; 36:875-887. [PMID: 30710230 DOI: 10.1007/s10815-019-01410-3] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2018] [Accepted: 01/18/2019] [Indexed: 12/29/2022] Open
Abstract
PURPOSE To investigate the association of cumulus cell (CC)-related expression of a selected cluster of key genes (PTGS2, CAMK1D, HAS2, STC1, and EFNB2) with embryo development to blastocyst. METHODS Exploratory study at a private clinic. Eighteen advanced maternal age patients were enrolled (37.3 ± 4.0 years). Seventy-five cumuli were collected, whose oocytes resulted in either developmental arrest (N = 33) or blastocyst formation (N = 42). The noninvasive CC gene expression was combined with time-lapse morphokinetic parameters and, for blastocysts, with qPCR-based aneuploidy testing on trophectoderm biopsies. RESULTS The detection rate was 100% for all transcripts, but STC1 (96%) and CAMK1D (89%). Among amplified assays, CC mean expression levels of CAMK1D, PTGS2, and HAS2 were higher from oocytes that developed to blastocyst. No difference in CC key gene expression was reported between euploid (N = 21) and aneuploid (N = 21) blastocysts. Some timings of early embryo development were faster in embryos developing to blastocyst (time of pronuclei appearance and fading, division to two- and four-cells, first and second cell cycles). However, the generalized linear models outlined increasing CAMK1D expression levels as the strongest parameter associated with oocytes' developmental potential from both a general (AUC = 0.78 among amplified samples) and an intrapatient perspectives (AUC = 0.9 among patients obtaining ≥ 2 zygotes from the cohort with different developmental outcomes). CONCLUSIONS CAMK1D level of expression in CCs associated with blastocyst development. If confirmed from larger studies in wider populations of patients, the investigation of CC key gene expression might suit IVF clinics not adopting blastocyst culture. Future investigations should clarify the role of CAMK1D in ovarian physiology and could provide novel insights on how oocytes gain competence during folliculogenesis.
Collapse
Affiliation(s)
- C Scarica
- DAHFMO, Unit of Histology and Medical Embryology, Sapienza, University of Rome, Rome, Italy.,Casa di cura Villa Salaria, Rome, Italy
| | - D Cimadomo
- Clinica Valle Giulia, G.EN.E.R.A. Centers for Reproductive Medicine, via G. De Notaris 2/b, Rome, Italy
| | - L Dovere
- Clinica Valle Giulia, G.EN.E.R.A. Centers for Reproductive Medicine, via G. De Notaris 2/b, Rome, Italy
| | - A Giancani
- DAHFMO, Unit of Histology and Medical Embryology, Sapienza, University of Rome, Rome, Italy.,Clinica Valle Giulia, G.EN.E.R.A. Centers for Reproductive Medicine, via G. De Notaris 2/b, Rome, Italy
| | - M Stoppa
- Clinica Valle Giulia, G.EN.E.R.A. Centers for Reproductive Medicine, via G. De Notaris 2/b, Rome, Italy
| | | | - F M Ubaldi
- Clinica Valle Giulia, G.EN.E.R.A. Centers for Reproductive Medicine, via G. De Notaris 2/b, Rome, Italy
| | - L Rienzi
- Clinica Valle Giulia, G.EN.E.R.A. Centers for Reproductive Medicine, via G. De Notaris 2/b, Rome, Italy.
| | - R Canipari
- DAHFMO, Unit of Histology and Medical Embryology, Sapienza, University of Rome, Rome, Italy
| |
Collapse
|
45
|
Daughtry BL, Rosenkrantz JL, Lazar NH, Fei SS, Redmayne N, Torkenczy KA, Adey A, Yan M, Gao L, Park B, Nevonen KA, Carbone L, Chavez SL. Single-cell sequencing of primate preimplantation embryos reveals chromosome elimination via cellular fragmentation and blastomere exclusion. Genome Res 2019; 29:367-382. [PMID: 30683754 PMCID: PMC6396419 DOI: 10.1101/gr.239830.118] [Citation(s) in RCA: 67] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2018] [Accepted: 01/22/2019] [Indexed: 12/12/2022]
Abstract
Aneuploidy that arises during meiosis and/or mitosis is a major contributor to early embryo loss. We previously showed that human preimplantation embryos encapsulate missegregated chromosomes into micronuclei while undergoing cellular fragmentation and that fragments can contain chromosomal material, but the source of this DNA was unknown. Here, we leveraged the use of a nonhuman primate model and single-cell DNA-sequencing (scDNA-seq) to examine the chromosomal content of 471 individual samples comprising 254 blastomeres, 42 polar bodies, and 175 cellular fragments from a large number (N = 50) of disassembled rhesus cleavage-stage embryos. Our analysis revealed that the aneuploidy and micronucleation frequency is conserved between humans and macaques, and that fragments encapsulate whole and/or partial chromosomes lost from blastomeres. Single-cell/fragment genotyping showed that these chromosome-containing cellular fragments (CCFs) can be maternally or paternally derived and display double-stranded DNA breaks. DNA breakage was further indicated by reciprocal subchromosomal losses/gains between blastomeres and large segmental errors primarily detected at the terminal ends of chromosomes. By combining time-lapse imaging with scDNA-seq, we determined that multipolar divisions at the zygote or two-cell stage were associated with CCFs and generated a random mixture of chromosomally normal and abnormal blastomeres with uniparental or biparental origins. Despite frequent chromosome missegregation at the cleavage-stage, we show that CCFs and nondividing aneuploid blastomeres showing extensive DNA damage are prevented from incorporation into blastocysts. These findings suggest that embryos respond to chromosomal errors by encapsulation into micronuclei, elimination via cellular fragmentation, and selection against highly aneuploid blastomeres to overcome chromosome instability during preimplantation development.
Collapse
Affiliation(s)
- Brittany L Daughtry
- Department of Cell, Developmental and Cancer Biology, Oregon Health and Science University School of Medicine, Portland, Oregon 97239, USA.,Division of Reproductive and Developmental Sciences, Oregon National Primate Research Center, Beaverton, Oregon 97006, USA
| | - Jimi L Rosenkrantz
- Division of Reproductive and Developmental Sciences, Oregon National Primate Research Center, Beaverton, Oregon 97006, USA.,Department of Molecular and Medical Genetics, Oregon Health and Science University School of Medicine, Portland, Oregon 97239, USA
| | - Nathan H Lazar
- Department of Medical Informatics and Clinical Epidemiology, Oregon Health and Science University School of Medicine, Portland, Oregon 97239, USA
| | - Suzanne S Fei
- Bioinformatics and Biostatistics Core, Oregon National Primate Research Center, Beaverton, Oregon 97006, USA
| | - Nash Redmayne
- Division of Reproductive and Developmental Sciences, Oregon National Primate Research Center, Beaverton, Oregon 97006, USA
| | - Kristof A Torkenczy
- Department of Molecular and Medical Genetics, Oregon Health and Science University School of Medicine, Portland, Oregon 97239, USA
| | - Andrew Adey
- Department of Molecular and Medical Genetics, Oregon Health and Science University School of Medicine, Portland, Oregon 97239, USA.,Department of Medicine, Knight Cardiovascular Institute, Oregon Health and Science University School of Medicine, Portland, Oregon 97239, USA
| | - Melissa Yan
- Bioinformatics and Biostatistics Core, Oregon National Primate Research Center, Beaverton, Oregon 97006, USA
| | - Lina Gao
- Bioinformatics and Biostatistics Core, Oregon National Primate Research Center, Beaverton, Oregon 97006, USA
| | - Byung Park
- Bioinformatics and Biostatistics Core, Oregon National Primate Research Center, Beaverton, Oregon 97006, USA
| | - Kimberly A Nevonen
- Department of Medicine, Knight Cardiovascular Institute, Oregon Health and Science University School of Medicine, Portland, Oregon 97239, USA
| | - Lucia Carbone
- Department of Molecular and Medical Genetics, Oregon Health and Science University School of Medicine, Portland, Oregon 97239, USA.,Department of Medical Informatics and Clinical Epidemiology, Oregon Health and Science University School of Medicine, Portland, Oregon 97239, USA.,Bioinformatics and Biostatistics Core, Oregon National Primate Research Center, Beaverton, Oregon 97006, USA.,Department of Medicine, Knight Cardiovascular Institute, Oregon Health and Science University School of Medicine, Portland, Oregon 97239, USA.,Division of Primate Genetics, Oregon National Primate Research Center, Beaverton, Oregon 97006, USA
| | - Shawn L Chavez
- Division of Reproductive and Developmental Sciences, Oregon National Primate Research Center, Beaverton, Oregon 97006, USA.,Department and Physiology and Pharmacology, Oregon Health and Science University School of Medicine, Portland, Oregon 97239, USA.,Department of Obstetrics and Gynecology, Oregon Health and Science University School of Medicine, Portland, Oregon 97239, USA.,Department of Biomedical Engineering, Oregon Health and Science University School of Medicine, Portland, Oregon 97239, USA
| |
Collapse
|
46
|
Albertini DF. Making sense out of syngamy at the onset of mammalian development. J Assist Reprod Genet 2018; 35:1357-1358. [PMID: 30069851 DOI: 10.1007/s10815-018-1282-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022] Open
|