Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 118
Filter
2.
Hum Reprod Open ; 2024(2): hoae023, 2024.
Article in English | MEDLINE | ID: mdl-38764910

ABSTRACT

STUDY QUESTION: Is there an association between morphokinetic variables of meiotic maturation and the severity of aneuploidy following in vitro maturation (IVM) in the mouse? SUMMARY ANSWER: The severity of meiotic aneuploidy correlates with an extended time to first polar body extrusion (tPB1) and duration of meiosis I (dMI). WHAT IS KNOWN ALREADY: Morphokinetic variables measured using time-lapse technology allow for the non-invasive evaluation of preimplantation embryo development within clinical assisted reproductive technology (ART). We recently applied this technology to monitor meiotic progression during IVM of mouse gametes. Whether there is a relationship between morphokinetic variables of meiotic progression and aneuploidy in the resulting egg has not been systematically examined at the resolution of specific chromosomes. Next-generation sequencing (NGS) is a robust clinical tool for determining aneuploidy status and has been reverse-translated in mouse blastocysts and oocytes. Therefore, we harnessed the technologies of time-lapse imaging and NGS to determine the relationship between the morphokinetics of meiotic progression and egg aneuploidy. STUDY DESIGN SIZE DURATION: Cumulus-oocyte complexes were collected from large antral follicles from hyperstimulated CD-1 mice. Cumulus cells were removed, and spontaneous IVM was performed in the absence or presence of two doses of Nocodazole (25 or 50 nM) to induce a spectrum of spindle abnormalities and chromosome segregation errors during oocyte meiosis. Comprehensive chromosome screening was then performed in the resulting eggs, and morphokinetic variables and ploidy status were compared across experimental groups (control, n = 11; 25 nM Nocodazole, n = 13; 50 nM Nocodazole, n = 23). PARTICIPANTS/MATERIALS SETTING METHODS: We monitored IVM in mouse oocytes using time-lapse microscopy for 16 h, and time to germinal vesicle breakdown (tGVBD), tPB1, and dMI were analyzed. Following IVM, comprehensive chromosome screening was performed on the eggs and their matched first polar bodies via adaptation of an NGS-based preimplantation genetic testing for aneuploidy (PGT-A) assay. Bioinformatics analysis was performed to align reads to the mouse genome and determine copy number-based predictions of aneuploidy. The concordance of each polar body-egg pair (reciprocal errors) was used to validate the results. Ploidy status was categorized as euploid, 1-3 chromosomal segregation errors, or ≥4 chromosomal segregation errors. Additionally, aneuploidy due to premature separation of sister chromatids (PSSC) versus non-disjunction (NDJ) was distinguished. MAIN RESULTS AND THE ROLE OF CHANCE: We applied and validated state-of-the-art NGS technology to screen aneuploidy in individual mouse eggs and matched polar bodies at the chromosome-specific level. By performing IVM in the presence of different doses of Nocodazole, we induced a range of aneuploidy. No aneuploidy was observed in the absence of Nocodazole (0/11), whereas IVM in the presence of 25 and 50 nM Nocodazole resulted in an aneuploidy incidence of 7.69% (1/13) and 82.61% (19/23), respectively. Of the aneuploid eggs, 5% (1/20) was due to PSSC, 65% (13/20) to NDJ, and the remainder to a combination of both. There was no relationship between ploidy status and tGVBD, but tPB1 and the dMI were both significantly prolonged in eggs with reciprocal aneuploidy events compared to the euploid eggs, and this scaled with the severity of aneuploidy. Eggs with ≥4 aneuploid chromosomes had the longest tPB1 and dMI (P < 0.0001), whereas eggs with one to three aneuploid chromosomes exhibited intermediate lengths of time (P < 0.0001). LARGE SCALE DATA: N/A. LIMITATIONS REASONS FOR CAUTION: We used Nocodazole in this study to disrupt the meiotic spindle and induce aneuploidy in mouse oocytes. Whether the association between morphokinetic variables of meiotic progression and the severity of aneuploidy occurs with other compounds that induce chromosome segregation errors remain to be investigated. In addition, unlike mouse oocytes, human IVM requires the presence of cumulus cells, which precludes visualization of morphokinetic variables of meiotic progression. Thus, our study may have limited direct clinical translatability. WIDER IMPLICATIONS OF THE FINDINGS: We validated NGS in mouse eggs to detect aneuploidy at a chromosome-specific resolution which greatly improves the utility of the mouse model. With a tractable and validated model system for characterizing meiotic aneuploidy, investigations into the molecular mechanisms and factors which may influence aneuploidy can be further elaborated. Time-lapse analyses of morphokinetic variables of meiotic progression may be a useful non-invasive predictor of aneuploidy severity. STUDY FUNDING/COMPETING INTERESTS: This work was supported by the Bill & Melinda Gates Foundation (INV-003385). Under the grant conditions of the Foundation, a Creative Commons Attribution 4.0 Generic License has already been assigned to the Author Accepted Manuscript version that might arise from this submission. The authors have no conflict of interest to disclose.

3.
J Assist Reprod Genet ; 41(1): 121-126, 2024 Jan.
Article in English | MEDLINE | ID: mdl-37957533

ABSTRACT

PURPOSE: This study aimed to evaluate whether a high-throughput high-resolution PGT-A method can detect copy number variants (CNVs) that could have clinical implications for patients and their embryos. METHODS: A prospective analysis of PGT-A cases was conducted using a high-resolution SNP microarray platform with over 820,000 probes. Cases where multiple embryos possessed the same segmental imbalance were identified, and preliminary PGT-A reports were issued recommending either parental microarray or conventional karyotyping to identify CNVs or translocations. RESULTS: Analysis of 6080 sequential PGT-A cases led to identification of 41 cases in which incidental findings were observed (0.7%) and parental testing was recommended. All cases, in which parental studies were completed, confirmed the original PGT-A incidental findings. In 2 of the cases, parental studies indicated a pathogenic variant with clinical implications for the associated embryos. In one of these cases, the patient was identified as a carrier of a duplication in chromosome 15q11.2:q11.2 (SNRPN + +), which is associated with autism spectrum disorder. In the second case, the patient was heterozygous positive for an interstitial deletion of 3p26.1:p26.3, which is associated with 3p deletion syndrome and had clinical implications for the patient and associated embryos. In each case, parental studies were concordant with PGT-A findings and revealed the presence of an otherwise unknown CNV. CONCLUSION: High-throughput high-resolution SNP array-based PGT-A has the ability to detect previously unknown and clinically significant parental deletions, duplications, and translocations. The use of cost-effective SNP array-based PGT-A methods may improve the effectiveness of PGT by identifying and preventing previously unknown pathogenic CNVs in children born to patients seeking in vitro fertilization.


Subject(s)
Chromosome Disorders , Preimplantation Diagnosis , Child , Female , Humans , Pregnancy , Aneuploidy , Chromosome Aberrations , Chromosome Disorders/diagnosis , DNA Copy Number Variations/genetics , Fertilization in Vitro , Genetic Testing/methods , Karyotyping , Preimplantation Diagnosis/methods , Translocation, Genetic/genetics
4.
F S Sci ; 4(2S): 1, 2023 05.
Article in English | MEDLINE | ID: mdl-37088300
5.
Am J Hum Genet ; 110(4): 565-574, 2023 04 06.
Article in English | MEDLINE | ID: mdl-36977411

ABSTRACT

Preimplantation genetic testing commonly employs simplistic copy-number analyses to screen for aneuploidy in blastocyst trophectoderm biopsies. Interpreting intermediate copy number alone as evidence of mosaicism has led to suboptimal estimation of its prevalence. Because mosaicism originates from mitotic nondisjunction, utilizing SNP microarray technology to identify the cell-division origins of aneuploidy might provide a more accurate estimation of its prevalence. The present study develops and validates a method of determining the cell-division origin of aneuploidy in the human blastocyst by using both genotyping and copy-number data in parallel. The concordance of predicted origins with expected results was demonstrated in a series of truth models (99%-100%). This included determination of X chromosome origins from a subset of normal male embryos, determination of the origins of translocation chromosome-related imbalances via embryos from couples with structural rearrangements, and prediction of either mitotic or meiotic origins via multiple rebiopsies of embryos with aneuploidy. In a cohort of blastocysts with parental DNA (n = 2,277), 71% were euploid, 27% were meiotic aneuploid, and 2% were mitotic aneuploid, indicating a low frequency of bona fide mosaicism in the human blastocyst (mean maternal age: 34.4). Chromosome-specific trisomies in the blastocyst were also consistent with observations previously established in products of conception. The ability to accurately identify mitotic-origin aneuploidy in the blastocyst could benefit and better inform individuals whose IVF cycle results in all aneuploid embryos. Clinical trials with this methodology might also help provide a definitive answer regarding the reproductive potential of bona fide mosaic embryos.


Subject(s)
Preimplantation Diagnosis , Pregnancy , Female , Humans , Male , Adult , Preimplantation Diagnosis/methods , Blastocyst , Aneuploidy , Genetic Testing/methods , Mosaicism
6.
F S Sci ; 4(2S): 27-35, 2023 05.
Article in English | MEDLINE | ID: mdl-36863445

ABSTRACT

OBJECTIVE: To validate the detection of abnormal ploidy in preimplantation embryos and evaluate its frequency in transferrable blastocysts. DESIGN: A high-throughput genome-wide single nucleotide polymorphism microarray-based preimplantation genetic testing (PGT) platform was validated using multiple positive controls, including cell lines of known haploid and triploid karyotypes and rebiopsies of embryos with initial abnormal ploidy results. This platform was then tested on all trophectoderm biopsies in a single PGT laboratory to calculate the frequency of abnormal ploidy and the parental and cell division origins of error. SETTING: Preimplantation genetic testing laboratory. PATIENT(S): The embryos from in vitro fertilization patients who elected for PGT were evaluated. Any patients who provided saliva samples were further analyzed for the parental and cell division origins of abnormal ploidy. INTERVENTION(S): None. MAIN OUTCOME MEASURE(S): Evaluable positive controls showed 100% concordance with original karyotypes. The overall frequency of abnormal ploidy within a single PGT laboratory cohort was 1.43%. RESULT(S): All cell lines showed 100% concordance with the expected karyotype. Additionally, all evaluable rebiopsies showed 100% concordance with the original abnormal ploidy karyotype. The frequency of abnormal ploidy was 1.43%, with 29% of those being haploid or uniparental isodiploid, 2.5% uniparental heterodiploid, 68% triploid, and 0.4% tetraploid. Twelve haploid embryos contained maternal deoxyribonucleic acid, and 3 contained paternal deoxyribonucleic acid. Thirty-four triploid embryos were of maternal origin, and 2 were of paternal origin. Thirty-five triploid embryos had a meiotic origin of error, and 1 was of mitotic error. Of those 35 embryos, 5 originated from meiosis I, 22 originated from meiosis II, and 8 were deemed inconclusive. On the basis of specific abnormal ploidy karyotypes, 41.2% of embryos would be falsely classified as euploid, and 22.7% would be false-positive mosaics with the use of the conventional next-generation sequencing-based PGT methods. CONCLUSION(S): This study demonstrates the validity of a high-throughput genome-wide single nucleotide polymorphism microarray-based PGT platform to accurately detect abnormal ploidy karyotypes and predict the parental and cell division origins of error of evaluable embryos. This unique method improves the sensitivity of detection for abnormal karyotypes, which can reduce the chances of adverse pregnancy outcomes.


Subject(s)
Preimplantation Diagnosis , Pregnancy , Female , Humans , Preimplantation Diagnosis/methods , Triploidy , Blastocyst/metabolism , Blastocyst/pathology , Ploidies , Abnormal Karyotype , DNA/metabolism
8.
Am J Hum Genet ; 109(9): 1572-1581, 2022 09 01.
Article in English | MEDLINE | ID: mdl-36055209

ABSTRACT

In IVF cycles, the application of aneuploidy testing at the blastocyst stage is quickly growing, and the latest reports estimate almost half of cycles in the US undergo preimplantation genetic testing for aneuploidies (PGT-A). Following PGT-A cycles, understanding the predictive value of an aneuploidy result is paramount for making informed decisions about the embryo's fate and utilization. Compelling evidence from non-selection trials strongly supports that embryos diagnosed with a uniform whole-chromosome aneuploidy very rarely result in the live birth of a healthy baby, while their transfer exposes women to significant risks of miscarriage and chromosomally abnormal pregnancy. On the other hand, embryos displaying low range mosaicism for whole chromosomes have shown reproductive capabilities somewhat equivalent to uniformly euploid embryos, and they have comparable clinical outcomes and gestational risks. Therefore, given their clearly distinct biological origin and clinical consequences, careful differentiation between uniform and mosaic aneuploidy is critical in both the clinical setting when counseling individuals and in the research setting when presenting aneuploidy studies in human embryology. Here, we focus on the evidence gathered so far on PGT-A diagnostic predictive values and reproductive outcomes observed across the broad spectrum of whole-chromosome aneuploidies detected at the blastocyst stage to obtain evidence-based conclusions on the clinical management of aneuploid embryos in the quickly growing PGT-A clinical setting.


Subject(s)
Preimplantation Diagnosis , Aneuploidy , Blastocyst , Female , Fertilization in Vitro , Genetic Testing , High-Throughput Nucleotide Sequencing , Humans , Live Birth , Mosaicism , Pregnancy
11.
Genes (Basel) ; 12(8)2021 07 21.
Article in English | MEDLINE | ID: mdl-34440279

ABSTRACT

Machine learning methods applied to large genomic datasets (such as those used in GWAS) have led to the creation of polygenic risk scores (PRSs) that can be used identify individuals who are at highly elevated risk for important disease conditions, such as coronary artery disease (CAD), diabetes, hypertension, breast cancer, and many more. PRSs have been validated in large population groups across multiple continents and are under evaluation for widespread clinical use in adult health. It has been shown that PRSs can be used to identify which of two individuals is at a lower disease risk, even when these two individuals are siblings from a shared family environment. The relative risk reduction (RRR) from choosing an embryo with a lower PRS (with respect to one chosen at random) can be quantified by using these sibling results. New technology for precise embryo genotyping allows more sophisticated preimplantation ranking with better results than the current method of selection that is based on morphology. We review the advances described above and discuss related ethical considerations.


Subject(s)
Embryo, Mammalian , Genetic Predisposition to Disease , Genetic Testing/ethics , Genetic Testing/methods , Multifactorial Inheritance , Humans
12.
Fertil Steril ; 116(5): 1205-1211, 2021 11.
Article in English | MEDLINE | ID: mdl-34304887

ABSTRACT

Preimplantation genetic testing for aneuploidy (PGT-A) remains one of the most controversial topics in reproductive medicine. With more than 40% of in vitro fertilization cycles in the United States reportedly involving PGT, both those in favor of and those opposed to PGT-A have significant interest in the efficacy of PGT-A. Ongoing issues include what patient population, if any, benefits from PGT-A, the true frequency of chromosomal mosaicism, whether embryonic aneuploidies self-correct, and how practitioners manage embryos designated as "mosaic." This review addresses several misconceptions and misinterpretations of data surrounding the genetic analysis and prediction of mosaicism in the preimplantation embryo.


Subject(s)
Blastocyst/pathology , Genetic Testing , Infertility/therapy , Mosaicism , Prenatal Diagnosis , Reproductive Techniques, Assisted/adverse effects , Aneuploidy , Embryo Transfer , Female , Genetic Counseling , Humans , Infertility/diagnosis , Infertility/physiopathology , Male , Predictive Value of Tests , Pregnancy , Reproducibility of Results , Risk Assessment , Risk Factors , Treatment Outcome
14.
F S Sci ; 2(3): 278-286, 2021 08.
Article in English | MEDLINE | ID: mdl-35560278

ABSTRACT

OBJECTIVE: To develop a test for evaluating the annexin A5 M2 haplotype in in vitro fertilization patients and preimplantation embryos. DESIGN: Test performance was measured by comparing Sanger sequencing of parental blood DNA and quantitative real-time polymerase chain reaction (qPCR) of saliva DNA, 3 fibroblast cell line 7-cell aliquots and their corresponding purified DNA, 123 trophectoderm biopsy samples, and DNA isolated from 1 embryonic stem cell line along with the Mendelian inheritance expectations, embryo Sanger sequencing, and single-nucleotide polymorphism (SNP) microarray-based linkage analysis. SETTING: Preimplantation genetic testing laboratory research on IVF patient and embryo DNA. PATIENT(S): An assay was developed for the detection of the M2 haplotype on saliva samples of 6 in vitro fertilization patients. In addition, 13 patients who underwent preimplantation genetic testing with data on parental and embryo biopsy DNA available for research use were evaluated. INTERVENTION(S): None. MAIN OUTCOME MEASURE(S): The concordance rates between Sanger sequencing, SNP array-based linkage analysis, and Mendelian inheritance expectations with qPCR. RESULT(S): The concordance rate between Sanger sequencing and qPCR was 100% on parental blood DNA and saliva DNA. The sample concordance rate between all replicates of 7-cell aliquots was 100%. The sample concordance rate between 3 cell lines used to prepare 7-cell aliquots and purified genomic DNA was 100%. The concordance rate between qPCR and Sanger sequencing results from a single trophectoderm biopsy and isolated embryonic stem cell line was 100%. The concordance rate of trophectoderm biopsy qPCR results and expectations from Mendelian inheritance rules was 97%; however, when SNP array-based linkage analysis was included, the concordance rate reached 100%. CONCLUSION(S): This study resulted in the development of a convenient saliva collection method and qPCR-based genotyping method to screen for the M2 haplotype. In addition, a novel method for testing preimplantation embryos has been established, providing an alternative to the use of low molecular weight heparin, through selection of embryos without the M2 haplotype.


Subject(s)
Preimplantation Diagnosis , Annexin A5/metabolism , Blastocyst/metabolism , DNA/metabolism , Female , Fertilization in Vitro , Haplotypes/genetics , Humans , Pregnancy , Preimplantation Diagnosis/methods
15.
Prenat Diagn ; 41(5): 545-553, 2021 04.
Article in English | MEDLINE | ID: mdl-32920823

ABSTRACT

Preimplantation genetic testing for aneuploidy (PGT-A) reduces miscarriage risk, increases the success of IVF, shortens time to pregnancy, and reduces multiple gestation rates without compromising outcomes. The progression of PGT-A has included common application of next-generation sequencing (NGS) from single nucleotide polymorphism microarray, quantitative real-time PCR, and array comparative hybridization platforms of analysis. Additional putative advances in PGT-A capability include classifying embryos as mosaic and predicting the presence of segmental imbalance. A critical component in the process of technical validation of these advancements involves evaluation of concordance between reanalysis results and initial testing results. While many independent studies have investigated the concordance of results obtained from the remaining embryo with the original PGT-A diagnosis, compilation and systematic analysis of published data has not been performed. Here, we review results from 26 primary research articles describing concordance in 1271 human blastocysts from 2260 pairwise comparisons. Results illustrate significantly higher discordance from PGT-A methods which utilize NGS and include prediction of mosaicism or segmental imbalance. These results suggest caution when considering new iterations PGT-A.


Subject(s)
Aneuploidy , Blastocyst/cytology , Preimplantation Diagnosis/trends , Adult , Blastocyst/physiology , Female , Humans , Pregnancy , Preimplantation Diagnosis/methods , Sensitivity and Specificity
16.
Cell ; 183(6): 1650-1664.e15, 2020 12 10.
Article in English | MEDLINE | ID: mdl-33125898

ABSTRACT

Correction of disease-causing mutations in human embryos holds the potential to reduce the burden of inherited genetic disorders and improve fertility treatments for couples with disease-causing mutations in lieu of embryo selection. Here, we evaluate repair outcomes of a Cas9-induced double-strand break (DSB) introduced on the paternal chromosome at the EYS locus, which carries a frameshift mutation causing blindness. We show that the most common repair outcome is microhomology-mediated end joining, which occurs during the first cell cycle in the zygote, leading to embryos with non-mosaic restoration of the reading frame. Notably, about half of the breaks remain unrepaired, resulting in an undetectable paternal allele and, after mitosis, loss of one or both chromosomal arms. Correspondingly, Cas9 off-target cleavage results in chromosomal losses and hemizygous indels because of cleavage of both alleles. These results demonstrate the ability to manipulate chromosome content and reveal significant challenges for mutation correction in human embryos.


Subject(s)
Alleles , CRISPR-Associated Protein 9/metabolism , Chromosomes, Human/genetics , Embryo, Mammalian/metabolism , Animals , Base Sequence , Blastocyst/metabolism , Cell Cycle/genetics , Cell Line , Chromosome Deletion , DNA Breaks, Double-Stranded , DNA End-Joining Repair/genetics , Embryo Implantation/genetics , Eye Proteins/genetics , Fertilization , Gene Editing , Gene Rearrangement/genetics , Genetic Loci , Genome, Human , Genotype , Heterozygote , Human Embryonic Stem Cells/metabolism , Humans , INDEL Mutation/genetics , Mice , Mitosis , Open Reading Frames/genetics , Polymorphism, Single Nucleotide/genetics
17.
Hum Reprod ; 35(9): 2134-2148, 2020 09 01.
Article in English | MEDLINE | ID: mdl-32772081

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.


Subject(s)
Aneuploidy , Exome , Adult , Animals , Blastocyst , Cell Cycle Proteins , Female , Humans , Mice , Middle Aged , Oocytes , Exome Sequencing , Young Adult
18.
Genes (Basel) ; 11(6)2020 06 12.
Article in English | MEDLINE | ID: mdl-32545548

ABSTRACT

Preimplantation genetic testing for polygenic disease risk (PGT-P) represents a new tool to aid in embryo selection. Previous studies demonstrated the ability to obtain necessary genotypes in the embryo with accuracy equivalent to in adults. When applied to select adult siblings with known type I diabetes status, a reduction in disease incidence of 45-72% compared to random selection was achieved. This study extends analysis to 11,883 sibling pairs to evaluate clinical utility of embryo selection with PGT-P. Results demonstrate simultaneous relative risk reduction of all diseases tested in parallel, which included diabetes, cancer, and heart disease, and indicate applicability beyond patients with a known family history of disease.


Subject(s)
Diabetes Mellitus, Type 1/diagnosis , Genetic Diseases, Inborn/diagnosis , Multifactorial Inheritance/genetics , Preimplantation Diagnosis/methods , Adult , Child, Preschool , Diabetes Mellitus, Type 1/genetics , Diabetes Mellitus, Type 1/pathology , Female , Genetic Diseases, Inborn/genetics , Genetic Diseases, Inborn/pathology , Humans , Male , Middle Aged , Pedigree , Risk Factors , Siblings
19.
Reproduction ; 160(5): A13-A17, 2020 Nov.
Article in English | MEDLINE | ID: mdl-32413844

ABSTRACT

Since its introduction to clinical practice, preimplantation genetic testing (PGT) has become a standard of care for couples at risk of having children with monogenic disease and for chromosomal aneuploidy to improve outcomes for patients with infertility. The primary objective of PGT is to reduce the risk of miscarriage and genetic disease and to improve the success of infertility treatment with the delivery of a healthy child. Until recently, the application of PGT to more common but complex polygenic disease was not possible, as the genetic contribution to polygenic disease has been difficult to determine, and the concept of embryo selection across multiple genetic loci has been difficult to comprehend. Several achievements, including the ability to obtain accurate, genome-wide genotypes of the human embryo and the development of population-level biobanks, have now made PGT for polygenic disease risk applicable in clinical practice. With the rapid advances in embryonic polygenic risk scoring, diverse considerations beyond technical capability have been introduced.


Subject(s)
Aneuploidy , Fertilization in Vitro/standards , Fetal Diseases/diagnosis , Genetic Diseases, Inborn/diagnosis , Genetic Testing/methods , Preimplantation Diagnosis/methods , Female , Fetal Diseases/genetics , Genetic Diseases, Inborn/embryology , Genetic Diseases, Inborn/genetics , Humans , Pregnancy
20.
F S Rep ; 1(3): 164-165, 2020 Dec.
Article in English | MEDLINE | ID: mdl-34223234
SELECTION OF CITATIONS
SEARCH DETAIL
...