Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 50
Filter
Add more filters

Country/Region as subject
Affiliation country
Publication year range
1.
Am J Hum Genet ; 108(3): 469-481, 2021 03 04.
Article in English | MEDLINE | ID: mdl-33626338

ABSTRACT

Total fertilization failure (TFF) can occur during in vitro fertilization (IVF) treatments, even following intracytoplasmic sperm injection (ICSI). Various male or female factors could contribute to TFF. Increasing evidence suggested that genetic variations in PLCZ1, which encodes 1-phosphatidylinositol 4,5-bisphosphate phosphodiesterase zeta-1 (PLCζ), is involved in oocyte activation and is a key male factor in TFF. In the present study, we explored the genetic variants in male individuals that led to TFF. A total of 54 couples with TFF or poor fertilization (fertilization rate < 20%) were screened, and 21 couples were determined to have a male infertility factor by the mouse oocyte activation test. Whole-exome sequencing of these 21 male individuals identified three homozygous pathogenic variants in ACTL9 (actin like 9) in three individuals. ACTL9 variations led to abnormal ultrastructure of the perinuclear theca (PT), and PLCζ was absent in the head and present in the neck of the mutant sperm, which contributed to failed normal calcium oscillations in oocytes and subsequent TFF. The key roles of ACTL9 in the PT structure and TFF after ICSI were further confirmed in an Actl9-mutated mouse model. Furthermore, assisted oocyte activation by calcium ionophore exposure successfully overcame TFF and achieved live births in a couple with an ACTL9 variant. These findings identified the role of ACTL9 in the PT structure and the correct localization of PLCζ. The results also provide a genetic marker and a therapeutic option for individuals who have undergone ICSI without successful fertilization.


Subject(s)
Actins/genetics , Infertility, Male/genetics , Phosphoinositide Phospholipase C/genetics , Spermatozoa/metabolism , Adult , Animals , Female , Fertilization in Vitro/adverse effects , Homozygote , Humans , Infertility, Male/pathology , Male , Mice , Oocytes/growth & development , Sperm Injections, Intracytoplasmic , Spermatozoa/pathology , Treatment Failure
2.
J Assist Reprod Genet ; 41(3): 739-750, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38263474

ABSTRACT

PURPOSE: The preimplantation genetic testing for aneuploidy (PGT-A) platform is not currently available for small copy-number variants (CNVs), especially those < 1 Mb. Through strategies used in PGT for monogenic disease (PGT-M), this study intended to perform PGT for families with small pathogenic CNVs. METHODS: Couples who carried small pathogenic CNVs and underwent PGT at the Reproductive and Genetic Hospital of CITIC-Xiangya (Hunan, China) between November 2019 and April 2023 were included in this study. Haplotype analysis was performed through two platforms (targeted sequencing and whole-genome arrays) to identify the unaffected embryos, which were subjected to transplantation. Prenatal diagnosis using amniotic fluid was performed during 18-20 weeks of pregnancy. RESULTS: PGT was successfully performed for 20 small CNVs (15 microdeletions and 5 microduplications) in 20 families. These CNVs distributed on chromosomes 1, 2, 6, 7, 13, 15, 16, and X with sizes ranging from 57 to 2120 kb. Three haplotyping-based PGT-M strategies were applied. A total of 89 embryos were identified in 25 PGT cycles for the 20 families. The diagnostic yield was 98.9% (88/89). Nineteen transfers were performed for 17 women, resulting in a 78.9% (15/19) clinical pregnancy rate after each transplantation. Of the nine women who had healthy babies, eight accepted prenatal diagnosis and the results showed no related pathogenic CNVs. CONCLUSION: Our results show that the extended haplotyping-based PGT-M strategy application for small pathogenic CNVs compensated for the insufficient resolution of PGT-A. These three PGT-M strategies could be applied to couples with small pathogenic CNVs.


Subject(s)
Abortion, Spontaneous , Preimplantation Diagnosis , Pregnancy , Humans , Female , Preimplantation Diagnosis/methods , Genetic Testing/methods , Pregnancy Rate , Abortion, Spontaneous/genetics , Live Birth , Aneuploidy
3.
Am J Hum Genet ; 107(1): 24-33, 2020 07 02.
Article in English | MEDLINE | ID: mdl-32502391

ABSTRACT

Zygotic cleavage failure (ZCF) is a unique early embryonic phenotype resulting in female infertility and recurrent failure of in vitro fertilization (IVF) and/or intracytoplasmic sperm injection (ICSI). With this phenotype, morphologically normal oocytes can be retrieved and successfully fertilized, but they fail to undergo cleavage. Until now, whether this phenotype has a Mendelian inheritance pattern and which underlying genetic factors play a role in its development remained to be elucidated. B cell translocation gene 4 (BTG4) is a key adaptor of the CCR4-NOT deadenylase complex, which is involved in maternal mRNA decay in mice, but no human diseases caused by mutations in BTG4 have previously been reported. Here, we identified four homozygous mutations in BTG4 (GenBank: NM_017589.4) that are responsible for the phenotype of ZCF, and we found they followed a recessive inheritance pattern. Three of them-c.73C>T (p.Gln25Ter), c.1A>G (p.?), and c.475_478del (p.Ile159LeufsTer15)-resulted in complete loss of full-length BTG4 protein. For c.166G>A (p.Ala56Thr), although the protein level and distribution of mutant BTG4 was not altered in zygotes from affected individuals or in HeLa cells, the interaction between BTG4 and CNOT7 was abolished. In vivo studies further demonstrated that the process of maternal mRNA decay was disrupted in the zygotes of the affected individuals, which provides a mechanistic explanation for the phenotype of ZCF. Thus, we provide evidence that ZCF is a Mendelian phenotype resulting from mutations in BTG4. These findings contribute to our understanding of the role of BTG4 in human early embryonic development and provide a genetic marker for female infertility.


Subject(s)
Cell Cycle Proteins/genetics , Infertility, Female/genetics , Mutation/genetics , Zygote/pathology , Animals , Cell Line, Tumor , Embryonic Development/genetics , Exoribonucleases/genetics , Female , HeLa Cells , Homozygote , Humans , Infertility, Female/pathology , Mice , Phenotype , RNA Stability/genetics
4.
J Transl Med ; 21(1): 779, 2023 11 02.
Article in English | MEDLINE | ID: mdl-37919732

ABSTRACT

BACKGROUND: Preimplantation genetic testing for aneuploidy (PGT-A) is widely used as an embryo selection technique in in vitro fertilization (IVF), but its effectiveness and potential beneficiary populations are unclear. METHODS: This retrospective cohort study included patients who underwent their first oocyte retrieval cycles at CITIC-Xiangya between January 2016 and November 2019, and the associated fresh and thawed embryo transfer cycles up to November 30, 2020. PGT-A (PGT-A group) and intracytoplasmic sperm injection (ICSI)/IVF (non-PGT-A group) cycles were included. The numbers of oocytes and embryos obtained were unrestricted. In total, 60,580 patients were enrolled, and baseline data were matched between groups using 1:3 propensity score matching. Sensitivity analyses, including propensity score stratification and traditional multivariate logistic regression, were performed on the original unmatched cohort to check the robustness of the overall results. Analyses were stratified by age, body mass index, ovarian reserve/responsiveness, and potential indications to explore benefits in subgroups. The primary outcome was cumulative live birth rate (CLBR). The other outcomes included live birth rate (LBR), pregnancy loss rate, clinical pregnancy rate, pregnancy complications, low birth weight rate, and neonatal malformation rate. RESULTS: In total, 4195 PGT-A users were matched with 10,140 non-PGT-A users. A significant reduction in CLBR was observed in women using PGT-A (27.5% vs. 31.1%; odds ratio (OR) = 0.84, 95% confidence interval (CI) 0.78-0.91; P < 0.001). However, women using PGT-A had higher first-transfer pregnancy (63.9% vs. 46.9%; OR = 2.01, 95% CI 1.81-2.23; P < 0.001) and LBR (52.6% vs. 34.2%, OR = 2.13, 95% CI 1.92-2.36; P < 0.001) rates and lower rates of early miscarriage (12.8% vs. 20.2%; OR = 0.58, 95% CI 0.48-0.70; P < 0.001), preterm birth (8.6% vs 17.3%; P < 0.001), and low birth weight (4.9% vs. 19.3%; P < 0.001). Moreover, subgroup analyses revealed that women aged ≥ 38 years, diagnosed with recurrent pregnancy loss or intrauterine adhesions benefited from PGT-A, with a significant increase in first-transfer LBR without a decrease in CLBR. CONCLUSION: PGT-A does not increase and decrease CLBR per oocyte retrieval cycle; nonetheless, it is effective in infertile populations with specific indications. PGT-A reduces complications associated with multiple gestations.


Subject(s)
Abortion, Spontaneous , Premature Birth , Pregnancy , Humans , Male , Infant, Newborn , Female , Oocyte Retrieval/methods , Retrospective Studies , Live Birth/epidemiology , Semen , Fertilization in Vitro/methods , Genetic Testing/methods , Abortion, Spontaneous/epidemiology , Aneuploidy
5.
J Med Genet ; 59(9): 850-857, 2022 Sep.
Article in English | MEDLINE | ID: mdl-34611029

ABSTRACT

BACKGROUND: Recurrent preimplantation embryo developmental arrest (RPEA) is the most common cause of assisted reproductive technology treatment failure associated with identified genetic abnormalities. Variants in known maternal genes can only account for 20%-30% of these cases. The underlying genetic causes for the other affected individuals remain unknown. METHODS: Whole exome sequencing was performed for 100 independent infertile females that experienced RPEA. Functional characterisations of the identified candidate disease-causative variants were validated by Sanger sequencing, bioinformatics and in vitro functional analyses, and single-cell RNA sequencing of zygotes. RESULTS: Biallelic variants in ZFP36L2 were associated with RPEA and the recurrent variant (p.Ser308_Ser310del) prevented maternal mRNA decay in zygotes and HeLa cells. CONCLUSION: These findings emphasise the relevance of the relationship between maternal mRNA decay and human preimplantation embryo development and highlight a novel gene potentially responsible for RPEA, which may facilitate genetic diagnoses.


Subject(s)
Infertility, Female , Blastocyst , Female , HeLa Cells , Humans , Infertility, Female/diagnosis , Infertility, Female/genetics , Pregnancy , RNA, Messenger, Stored , Transcription Factors/genetics , Exome Sequencing
6.
J Assist Reprod Genet ; 40(11): 2609-2618, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37728792

ABSTRACT

PURPOSE: We aimed to compare embryo development, cumulative live birth rate (CLBR), and perinatal outcomes of embryos cultured in 20% and 5% oxygen from days 1 to 3 after insemination. METHODS: This retrospective study included patients who received in vitro fertilization (IVF) treatment between January 2015 and November 2019. Embryos of each patient were cultured at 20% or 5% oxygen from days 1-3 after insemination. The primary outcome was CLBR. Propensity score matching (PSM) was used to balance patients' baseline data in both oxygen groups. RESULTS: In total, 31,566 patients were enrolled. After PSM, the rate of high-quality day 3 embryos was significantly lower in the 20% than in the 5% oxygen group (0.49 ± 0.33 vs 0.51 ± 0.33; adjusted ß = -0.03; 95% confidence interval [CI], -0.03 to -0.02). The CLBR was significantly lower in the 20% than in the 5% oxygen group (58.6% vs. 62.4%; adjusted odds ratio = 0.85; 95% CI, 0.81-0.90). The birthweight and Z score of singletons were significantly higher in the 20% than in the 5% oxygen group (birthweight: 3.30 ± 0.50 vs. 3.28 ± 0.48; adjusted ß = 0.022; 95% CI, 0.004-0.040; Z score: 0.26 ± 1.04 vs. 0.22 ± 1.01; adjusted ß = 0.037; 95% CI, 0.001-0.074). CONCLUSION: Culturing embryos at atmospheric oxygen concentrations from days 1 to 3 compromises embryo quality, reduces CLBR, and affects birthweight. The 5% oxygen concentration is more suitable for embryo culture in IVF laboratories to achieve successful outcomes.


Subject(s)
Birth Rate , Fertilization in Vitro , Pregnancy , Female , Humans , Birth Weight , Retrospective Studies , Insemination , Live Birth/epidemiology , Pregnancy Rate
7.
J Assist Reprod Genet ; 40(8): 1949-1959, 2023 Aug.
Article in English | MEDLINE | ID: mdl-37428430

ABSTRACT

PURPOSE: To evaluate whether outcomes of in vitro fertilization (IVF) are affected during the coronavirus disease-19 (COVID-19) pandemic. METHODS: This was a single-center, retrospective study. Embryo development, pregnancy, and live birth outcomes were compared between COVID-19 and pre-COVID-19 groups. Blood samples from patients during the COVID-19 pandemic were tested for COVID-19. RESULTS: After 1:1 random matching, 403 cycles for each group were included in the study. The rates of fertilization, normal fertilization, and blastocyst formation were higher in the COVID-19 group than in the pre-COVID-19 group. No difference was observed in the rates of day 3 good-quality embryos and good-quality blastocysts between the groups. A multivariate analysis showed that the live birth rate in the COVID-19 group was higher than that in the pre-COVID-19 group (51.4% vs. 41.4%, P = 0.010). In fresh cleavage-stage embryo and blastocyst transfer cycles, there were no differences between the groups in terms of pregnancy, obstetric, and perinatal outcomes. In the freeze-all cycles, the live birth rate was higher during the COVID-19 pandemic (58.0% vs. 34.5%, P = 0.006) than during the pre-COVID-19 period following frozen cleavage stage embryo transfer. The rate of gestational diabetes during the COVID-19 pandemic was higher than that during the pre-COVID-19 period (20.3% vs. 2.4%, P = 0.008) following frozen blastocyst transfer. All the serological results of the patients during the COVID-19 pandemic were negative. CONCLUSION: Our results indicate that embryo development, pregnancy, and live birth outcomes in uninfected patients were not compromised during the COVID-19 pandemic at our center.


Subject(s)
COVID-19 , Pandemics , Pregnancy , Female , Humans , Live Birth/epidemiology , Retrospective Studies , COVID-19/epidemiology , Fertilization in Vitro , Embryonic Development , Birth Rate , Pregnancy Rate , Blastocyst
8.
J Assist Reprod Genet ; 40(10): 2333-2342, 2023 Oct.
Article in English | MEDLINE | ID: mdl-37656381

ABSTRACT

PURPOSE: To investigate the feasibility of the application of conventional in vitro fertilization (cIVF) for couples undergoing preimplantation genetic testing for aneuploidies (PGT-A) with non-male factor infertility. METHODS: To evaluate the efficiency of sperm whole-genome amplification (WGA), spermatozoa were subjected to three WGA protocols: Picoplex, ChromInst, and multiple displacement amplification (MDA). In the clinical studies, 641 couples who underwent PGT-A treatment for frozen embryos between January 2016 and December 2021 were included to retrospectively compare the chromosomal and clinical outcomes of cIVF and intracytoplasmic sperm injection (ICSI). Twenty-six couples were prospectively recruited for cIVF and PGT-A treatment between April 2021 and April 2022; parental contamination was analyzed in biopsied samples; and 12 aneuploid embryos were donated to validate the PGT-A results. RESULTS: Sperm DNA failed to amplify under Picoplex and ChromInst conditions but could be amplified using MDA. In frozen PGT-A cycles, no significant differences in the average rates of euploid, mosaic, and aneuploid embryos per cycle between the cIVF-PGT-A and ICSI-PGT-A groups were observed. The results of the prospective study that recruited couples for cIVF-PGT-A treatment showed no paternal contamination and one case of maternal contamination in 150 biopsied trophectoderm samples. Among the 12 donated embryos with whole-chromosome aneuploidy, 11 (91.7%) presented uniform chromosomal aberrations, which were in agreement with the original biopsy results. CONCLUSIONS: Under the Picoplex and ChromInst WGA protocols, the risk of parental contamination in the cIVF-PGT-A cycles was low. Therefore, applying cIVF to couples with non-male factor infertility who are undergoing PGT-A is feasible.


Subject(s)
Infertility , Semen , Humans , Male , Prospective Studies , Retrospective Studies , Aneuploidy , Fertilization in Vitro , Genetic Testing
9.
J Transl Med ; 20(1): 396, 2022 09 04.
Article in English | MEDLINE | ID: mdl-36058949

ABSTRACT

BACKGROUND: Previous studies suggested that non-invasive preimplantation genetic testing (niPGT) for intracytoplasmic sperm injection (ICSI) blastocysts can be used to identify chromosomal ploidy and chromosomal abnormalities. Here, we report the feasibility and performance of niPGT for conventional in vitro fertilization (IVF) blastocysts. METHODS: This was a prospective observational study. In the preclinical stage, whole genome amplification and NGS were performed using the sperm spent culture medium (SCM). Then, trophectoderm (TE) biopsies and corresponding SCM derived from 27 conventional IVF monopronuclear embryos were collected. In the clinical stage, samples from 25 conventional IVF cycles and 37 ICSI cycles from April 2020-August 2021 were collected for performance evaluation. RESULTS: Preclinically, we confirmed failed sperm DNA amplification under the current amplification system. Subsequent niPGT from the 27 monopronuclear blastocysts showed 69.2% concordance with PGT results of corresponding TE biopsies. In the clinical stage, no paternal contamination was observed in any of the 161 SCM samples from conventional IVF. While maternal contamination was observed in 29.8% (48/161) SCM samples, only 2.5% (4/161) samples had a contamination ratio ≥ 50%. Compared with that of TE biopsy, the performances of NiPGT from 161 conventional IVF embryos and 122 ICSI embryos were not significantly different (P > 0.05), with ploidy concordance rates of 75% and 74.6% for IVF and ICSI methods, respectively. Finally, evaluation of the euploid probability of embryos with different types of niPGT results showed prediction probabilities of 82.8%, 77.8%, 62.5%, 50.0%, 40.9% and 18.4% for euploidy, sex-chromosome mosaics only, low-level mosaics, multiple abnormal chromosomes, high-level mosaics and aneuploidy, respectively. CONCLUSIONS: Our research results preliminarily confirm that the niPGT approach using SCM from conventional IVF has comparable performance with ICSI and might broadening the application scope of niPGT.


Subject(s)
Preimplantation Diagnosis , Blastocyst/pathology , Chromosome Aberrations , Culture Media , Female , Fertilization in Vitro , Genetic Testing/methods , Humans , Male , Pregnancy , Preimplantation Diagnosis/methods , Semen
10.
Genet Med ; 24(11): 2285-2295, 2022 11.
Article in English | MEDLINE | ID: mdl-36107168

ABSTRACT

PURPOSE: This study aimed to investigate the spectrum and characteristics of segmental aneuploidies (SAs) of <10 megabase (Mb) length in human preimplantation blastocysts. METHODS: Preimplantation genetic testing for aneuploidy was performed in 15,411 blastocysts from 5171 patients using a validated 1 Mb resolution platform. The characteristics and spectrum of SAs, including the incidence, sizes, type, inheritance pattern, clinical significance, and embryo distribution, were studied. RESULTS: In total, 6.4% of the 15,411 blastocysts carried SAs of >10 Mb, 4.9% of embryos had SAs ranging between 1 to 10 Mb, and 84.3% of 1 to 10 Mb SAs were <5 Mb in size. Inheritance pattern analysis indicated that approximately 63.8% of 1 to 10 Mb SAs were inherited and were predominantly 1 to 3 Mb in size. Furthermore, 18.4% of inherited SAs and 51.9% de novo 1 to 10 Mb SAs were pathogenic or likely pathogenic (P/LP). Different from whole-chromosome aneuploidies, reanalysis indicated that 50% of the de novo 1 to 10 Mb SAs and 70% of the >10 Mb SAs arose from mitotic errors. CONCLUSION: Based on the established platform, 1 to 10 Mb SAs are common in blastocysts and include a subset of P/LP SAs. Inheritance pattern analysis and clinical interpretation based on the American College of Medical Genetics and Genomics/Association for Molecular Pathology guidelines contributed to determine the P/LP SAs.


Subject(s)
Preimplantation Diagnosis , Pregnancy , Female , Humans , Aneuploidy , Blastocyst , Genetic Testing
11.
BMC Biol ; 19(1): 200, 2021 09 09.
Article in English | MEDLINE | ID: mdl-34503495

ABSTRACT

BACKGROUND: Although oocyte quality is the dominant factor determining embryo quality, few studies have been conducted to evaluate embryo quality based on the metabolites related to the oocyte. With quantification of the follicular fluid (FF) metabolites, in assisted reproductive technology (ART), this study sought to evaluate the embryo or oocyte quality through an informative approach. RESULTS: An evaluation model consisting of 17 features was generated to distinguish the embryo quality on day 3 post-fertilization, and phosphatidylcholines (PCs) were the key contributors to the evaluation. The model was extended to the patients under different ages and hyperstimulations, and the features were further enriched to facilitate the evaluation of the embryo quality. The metabolites were clustered through pathway analysis, leading to a hypothesis that accumulation of arachidonic acid induced by PCs might weaken embryo quality on day 3 post-fertilization. CONCLUSIONS: A discriminating model with metabolic features elicited from follicular fluid was established, which enabled the evaluation of the embryo or oocyte quality even under certain clinical conditions, and the increase of PCs in follicular fluid implies the attenuation of embryo quality on day 3 post-fertilization.


Subject(s)
Embryonic Development , Follicular Fluid , Phosphatidylcholines , Female , Fertilization , Fertilization in Vitro , Humans , Oocytes
12.
Clin Genet ; 99(2): 286-291, 2021 02.
Article in English | MEDLINE | ID: mdl-33020905

ABSTRACT

The subcortical maternal complex (SCMC) is an oocyte-to-embryo-specific maternal functional module. Some variants of SCMC genes that contribute to preimplantation embryonic arrest have been identified. However, more novel variants should be identified to broaden the genetic and phenotypic spectrum of SCMC genes and establish their roles in embryonic development. We identified 13 novel variants in the SCMC genes, TLE6, NLRP5, NLRP2, and PADI6, from 10 of a total of 50 infertile females with recurrent preimplantation embryonic arrest. Six variants in TLE6 were found in five patients with embryonic arrest, accompanied by direct cleavage and severe fragmentation at the cleavage stage. Three patients carried NLRP5 variants, and one patient each who carried NLRP2 and PADI6 variants had subsequent poor or failed fertilization and cleavage arrest with a relatively lower ratio of severely fragmented embryos. Our findings expand the genetic and phenotypic spectrum of SCMC genes associated with human embryogenesis and might help lay the foundation for the genetic diagnosis of female infertility.


Subject(s)
Abortion, Spontaneous/genetics , Blastocyst , Infertility, Female/genetics , Multiprotein Complexes/genetics , Adaptor Proteins, Signal Transducing/genetics , Apoptosis Regulatory Proteins/genetics , Autoantigens/genetics , Co-Repressor Proteins/genetics , Embryonic Development/genetics , Female , Genetic Variation , Humans , Mitochondrial Proteins/genetics , Nuclear Proteins/genetics , Pedigree , Phenotype , Protein-Arginine Deiminase Type 6/genetics , Time-Lapse Imaging , Whole Genome Sequencing
13.
Reprod Biomed Online ; 43(2): 215-222, 2021 Aug.
Article in English | MEDLINE | ID: mdl-34193357

ABSTRACT

RESEARCH QUESTION: What is the incidence of complex mosaic in preimplantation genetic testing (PGT) blastocysts and can it be managed in clinical practice? DESIGN: A retrospective study of PGT cycles conducted between January 2018 and October 2019 at a single centre. Biopsies of blastocysts were collected and analysed by next-generation sequencing (NGS). Complex mosaic blastocysts were defined as those with three or more mosaic chromosomes. The cryopreserved complex mosaic blastocysts underwent a second round of biopsy, NGS analysis and vitrification. The euploid blastocysts identified by the re-biopsy were warmed again for embryo transfer. The main outcomes included the prevalence of the complex mosaic and the ongoing pregnancy rate. RESULTS: The prevalence of the complex mosaic was 2.4% (437/17,979). The prevalence of the complex mosaic was not associated with maternal age and morphological quality. A total of 89 complex mosaic blastocysts underwent re-biopsy and 96.6% (86/89) survived the first warming. For the re-biopsy samples, 61.6% (53/86) were euploid. The poor-quality blastocysts had higher rates of aneuploidy compared with good-quality blastocysts. The survival rate for blastocysts undergoing the second warming was 100% (18/18) and resulted in an ongoing pregnancy rate of 38.9% (7/18) as well as the birth of six healthy infants. CONCLUSION: Re-biopsy may rescue blastocysts with development potential for transfer and improve the cumulative pregnancy rate per stimulation cycle in patients containing complex mosaic blastocysts.


Subject(s)
Blastocyst/pathology , Infertility/diagnosis , Mosaicism , Adult , Biopsy , Blastocyst/metabolism , Chromosome Aberrations/embryology , Chromosome Aberrations/statistics & numerical data , Cryopreservation , Embryo Transfer/statistics & numerical data , Female , High-Throughput Nucleotide Sequencing , Humans , Infertility/epidemiology , Infertility/genetics , Infertility/therapy , Mosaicism/embryology , Mosaicism/statistics & numerical data , Pregnancy , Pregnancy Rate , Preimplantation Diagnosis/statistics & numerical data , Prevalence , Prognosis , Retrospective Studies , Treatment Outcome , Vitrification
14.
J Assist Reprod Genet ; 38(9): 2261-2272, 2021 Sep.
Article in English | MEDLINE | ID: mdl-33970371

ABSTRACT

PURPOSE: TUBB8 is a gene that is frequently analysed in the genetic diagnosis of female infertility; 102 variants of this gene have been identified. However, the evaluation of its pathogenicity and the resulting phenotypes vary. Here, we aimed to identify novel TUBB8 variants as well as to summarize the reported variants and phenotypes in order for them to be included in genetic counselling analyses. METHODS: We performed whole exome sequencing to screen for candidate variants in 100 infertile female subjects and 100 controls who were able to conceive naturally. All variants were confirmed by Sanger sequencing. The effects of the variants in oocytes/arrested embryos were assessed by morphological observations, polar body biopsies, and chromosome analysis. A molecular modelling analysis was used to evaluate the possible effects of variants on protein secondary structure. RESULTS: We identified 29 TUBB8 variants, of which 20 were novel and five were maternally inherited. We identified three of a total of six recurrent variants that were specific for complete cleavage failure. Moreover, we obtained evidence that TUBB8 variants with large polar bodies had chromosome segregation errors. CONCLUSIONS: Our study expands the spectrum of TUBB8 variants, particularly for embryonic arrest. Together with the extant knowledge of TUBB8 variants, this study provides a foundation for the genetic counselling of female infertility.


Subject(s)
Infertility, Female/pathology , Mutation , Phenotype , Tubulin/genetics , Female , Humans , Infertility, Female/genetics
15.
Hum Reprod ; 35(5): 1120-1129, 2020 05 01.
Article in English | MEDLINE | ID: mdl-32358599

ABSTRACT

STUDY QUESTION: Is the mitochondrial DNA (mtDNA) copy number of cumulus granulosa cells (CGCs) related to the maturation of oocyte cytoplasm? SUMMARY ANSWER: Compared with the mtDNA copy number of CGCs from germinal vesicles (GV), CGCs from Metaphase I (MI) oocytes appear to have a lower mtDNA copy number. WHAT IS KNOWN ALREADY: The growth and development of CGCs and oocyte are synchronised. The interaction between CGCs and the oocyte provides the appropriate balance of energy, which is necessary for mammalian oocyte development. Moreover, in the oocyte-cumulus complex (OCC), mature oocytes with higher mtDNA copy numbers tend to have corresponding CGCs with higher mtDNA copy numbers. STUDY DESIGN, SIZE, DURATION: This is a prospective study of 302 OCCs obtained from 70 women undergoing in vitro fertilisation with intracytoplasmic sperm injection (ICSI) at the Reproductive and Genetic Hospital of CITIC-Xiangya, between 24 February 2018 and 21 December 2019. The CGCs were divided into three groups (GV, MI and MII stages) based on the maturation status of their corresponding oocyte. The sample sizes (n = 302) of CGCs in the three stages were 63 (CGCGV), 70 (CGCMI) and 169 (CGCMII), respectively. Some of the samples (n = 257) was used to quantify the mtDNA copy number, while the rest (n = 45) were used to analyse the expression level of mitochondrial genes. Furthermore, we retrieved 82 immature oocytes from among the 257 OCCs used for mtDNA copy numbers, including 36 GV oocytes and 46 MI oocytes, for analysis of oocyte mtDNA. PARTICIPANTS/MATERIALS, SETTING, METHODS: We selected genes with high consistency of real-time PCR results to accurately measure the mtDNA copy number by testing the efficacy and the reproducibility in whole genome amplification (WGA) samples from a human embryonic stem cell line. The CGCs of each oocyte were individually isolated. The mtDNA copy number and gene expression of the CGCs were assessed using real-time PCR techniques. Mitochondrial DNA copy number of the corresponding immature oocytes was also evaluated. MAIN RESULTS AND THE ROLE OF CHANCE: MT-ND1, MT-CO1 and ß-globin genes were chosen for the assessment of mtDNA content, and mRNA expressions of MT-ND1, MT-CO1, PGC-1α and TFAM were also measured. The genome of 257 CGCs and 82 immature oocytes were amplified according to the multiple displacement amplification (MDA) protocol, and RNA was extracted from 45 CGCs. Compared with CGCGV, CGCMI had a significantly lower mtDNA copy number. In the MT-ND1 assay, the CGCGV: CGCMI was [270 ± 302]: [134 ± 201], P = 0.015. In the MT-CO1 assay, CGCGV: CGCMI was [205 ± 228]: [92 ± 112], P = 0.026. There was no statistical difference in mtDNA between CGCGV and CGCMII. In the MT-ND1 assay, CGCGV: CGCMII was [270 ± 302]: [175 ± 223], P = 0.074. In the MT-CO1 assay, CGCGV: CGCMII was [205 ± 228]: [119 ± 192], P = 0.077. No statistical difference of mtDNA copy number was observed between CGCMI and CGCMII. In the MT-ND1 assay, CGCMI: CGCMII was [134 ± 201]: [175 ± 223], P = 0.422. In the MT-CO1 assay, CGCMI: CGCMII was [92 ± 112]: [119 ± 192], P = 0.478. To verify the reliability of the above results, we further analysed the mtDNA copy number of CGCs of 14 patients with GV, MI and MII oocytes, and the results showed that the mtDNA copy number of CGCMI may be lower. The mtDNA copy number of CGCGV and CGCMI was statistically different in the MT-ND1 assay where CGCGV: CGCMI was [249 ± 173]: [118 ± 113], P = 0.016, but in the MT-CO1 assay, CGCGV: CGCMI was [208 ± 199]: [83 ± 98], P = 0.109. There was no significant difference in mtDNA between CGCGV and CGCMII. In the MT-ND1 assay, CGCGV: CGCMII was [249 ± 173]: [185 ± 200], P = 0.096. In the MT-CO1 assay, CGCGV: CGCMII was [208 ± 199]: [114 ± 139], P = 0.096. There was also no significant difference in mtDNA between CGCMI and CGCMII. In the MT-ND1 assay, CGCMI: CGCMII was [118 ± 113]: [185 ± 200], P = 0.198. In the MT-CO1 assay, CGCMI: CGCMII was [83 ± 98]: [114 ± 139], P = 0.470. Moreover, there were no statistical differences in the expression levels of MT-ND1, MT-CO1, PGC-1α and TFAM between CGCGV, CGCMI and CGCMII (P > 0.05). LARGE SCALE DATA: N/A. LIMITATIONS, REASONS FOR CAUTION: Due to the ethical issues, the study did not quantify the mtDNA content of MII oocytes. Thus, whether the change in mtDNA copy number in CGCs is related to the different developmental stages of oocytes has not been further confirmed. Moreover, the sample size was relatively small. WIDER IMPLICATIONS OF THE FINDINGS: The mtDNA copy number of CGCs decreases from the GV phase to the MI phase and stays steady from the MI to MII stage. At different stages of oocyte maturation, the mtDNA of CGCs may undergo self-degradation and replication to meet the energy requirements of the corresponding oocyte and the maturation of the oocyte cytoplasm. STUDY FUNDING/COMPETING INTEREST(S): Funding was provided by the National Key R&D Program of China (Grant 2018YFC1003100, to L.H.), the science and technology major project of the Ministry of Science and Technology of Hunan Province, China (grant 2017SK1030, to G.L.), the National Natural Science Foundation of China (grant 81873478, to L.H.), and Merck Serono China Research Fund for Fertility Experts (to L.H.). There is no conflict of interest.


Subject(s)
Cumulus Cells , DNA, Mitochondrial , Animals , China , DNA Copy Number Variations , DNA, Mitochondrial/genetics , Female , Humans , In Vitro Oocyte Maturation Techniques , Oocytes , Prospective Studies , Reproducibility of Results
16.
Genet Med ; 21(2): 431-440, 2019 02.
Article in English | MEDLINE | ID: mdl-29895852

ABSTRACT

PURPOSE: The oocyte-borne genetic causes leading to fertilization failure are largely unknown. We aimed to identify novel human pathogenic variants (PV) and genes causing fertilization failure. METHODS: We performed exome sequencing for a consanguineous family with a recessive inheritance pattern of female infertility characterized by oocytes with a thin zona pellucida (ZP) and fertilization failure in routine in vitro fertilization. Subsequent PV screening of ZP2 was performed in additional eight unrelated infertile women whose oocytes exhibited abnormal ZP and similar fertilization failure. Expression of ZP proteins was assessed in mutant oocytes by immunostaining, and functional studies of the wild-type and mutant proteins were carried out in CHO-K1 cells. RESULTS: Two homozygous s PV (c.1695-2A>G, and c.1691_1694dup (p.C566Wfs*5), respectively) of ZP2 were identified in the affected women from two unrelated consanguineous families. All oocytes carrying PV were surrounded by a thin ZP that was defective for sperm-binding. Immunostaining indicated a lack of ZP2 protein in the thin ZP. Studies in CHO cells showed that both PV resulted in a truncated ZP2 protein, which might be intracellularly sequestered and prematurely interacted with other ZP proteins. CONCLUSION: We identified loss-of-function PV of ZP2 causing a structurally abnormal and dysfunctional ZP, resulting in fertilization failure and female infertility.


Subject(s)
Fertilization in Vitro , Infertility, Female/genetics , Zona Pellucida Glycoproteins/genetics , Adult , Animals , CHO Cells , Cricetulus , Female , Humans , Infertility, Female/pathology , Infertility, Female/physiopathology , Mutation , Pregnancy , Sequence Analysis, DNA , Treatment Failure , Zona Pellucida/ultrastructure , Zona Pellucida Glycoproteins/metabolism
17.
Hum Reprod ; 34(11): 2201-2207, 2019 11 01.
Article in English | MEDLINE | ID: mdl-31734689

ABSTRACT

Empty follicle syndrome (EFS) is the complete failure to retrieve oocytes after ovarian stimulation. Although LHCGR and ZP3 were identified as causative genes, it is still unclear what happens to these patients' oocytes, and the pathogenesis of EFS remains obscure. Here, we identified six novel ZP1 mutations associated with EFS and female infertility that was inherited recessively in five unrelated families. Studies in CHO-K1 cells showed that these mutations resulted in either degradation or truncation of ZP1 protein. Immunohistochemistry using ovarian serial sections demonstrated that all preantral follicles had normal architecture, but with a thin ZP, lacking ZP1, surrounding the growing oocytes. The antral follicles were also defective in normal cumulus-oocyte complex organisation, leading us to speculate that the lack of ZP1 might lead to oocyte degeneration or increased fragility of the oocyte during follicular puncture, ultimately resulting in EFS. To our knowledge, this is the first study that presents morphological evidence showing normal preantral folliculogenesis with abnormal ZP assembly in EFS patients. Our data provides a better understanding of the biological functions of ZP1 in human ZP assembly and folliculogenesis and gives new insights into the pathogenesis of EFS and possible therapeutic developments.


Subject(s)
Oocytes/cytology , Ovarian Diseases/genetics , Ovarian Follicle/pathology , Zona Pellucida Glycoproteins/genetics , Zona Pellucida/pathology , Adult , Alleles , Exome , Female , Genotype , Humans , Infertility, Female , Ovarian Reserve , Ovulation Induction , Sequence Analysis, DNA
18.
J Assist Reprod Genet ; 36(5): 925-933, 2019 May.
Article in English | MEDLINE | ID: mdl-30924053

ABSTRACT

PURPOSE: This study aimed to clarify the risks of adverse pregnancy outcomes in patients and their offspring after frozen embryo transfer (FET) during an artificial cycle (AC). METHODS: We conducted a retrospective cohort study that included all FET cycles and subsequent deliveries in a single centre between August 2013 and March 2016. Pregnancy, obstetric and neonatal outcomes were compared among patients treated during an AC or a natural cycle with luteal phase support (NC-LPS). Multivariate logistic regression was performed to evaluate the relationship between endometrial preparation schemes and pregnancy, obstetric and neonatal outcomes. RESULTS: AC-FET was not a significant risk factor for clinical pregnancy rate, multiple birth rate or miscarriage rate after adjusting for potential confounders. However, AC-FET was a significant risk factor for ectopic pregnancy rate (adjusted odds ratio (AOR), 1.738; 95% confidence interval (CI), 1.086-2.781) and live birth rate (AOR, 0.709; 95% CI, 0.626-0.802). Regarding obstetric outcomes, AC-FET was found to be associated with an increased risk for hypertension disorder (AOR, 1.780; 95% CI, 1.262-2.510) and caesarean section (AOR, 1.507; 95% CI, 1.195-1.900). In multiples, birth weight (2550 g (2150-2900 g) in AC-FET vs. 2600 g (2350-2900 g) in NC-LPS; P = 0.023), gestational age (36.6 weeks (35.3-37.6 weeks) vs. 37.1 weeks (36.1-37.9 weeks); P < 0.001), and z-score (- 0.5 (- 1.1, - 0.0) vs. - 0.4 (- 1.0, 0.2); P = 0.009) were higher in the NC-LPS group than in the AC-FET group, although there were no differences in these variables among singletons. CONCLUSION: Compared with NC-LPS, AC-FET seemed to have a negative effect on obstetric outcomes.


Subject(s)
Cesarean Section/statistics & numerical data , Cryopreservation/methods , Embryo Transfer/adverse effects , Fertilization in Vitro/methods , Pregnancy Complications/etiology , Pregnancy Rate , Premature Birth/etiology , Adult , Female , Humans , Infant, Newborn , Pregnancy , Pregnancy Outcome , Retrospective Studies
19.
Arch Gynecol Obstet ; 300(5): 1227-1237, 2019 11.
Article in English | MEDLINE | ID: mdl-31552485

ABSTRACT

PURPOSE: To determine whether the endometrial thickness (EMT) affects the occurrence of obstetric complications and neonatal outcomes in frozen embryo transfer (FET). METHODS: We conducted a retrospective study that included singleton deliveries (N = 5251) resulting from FET in a single center between August 2013 and March 2016. Obstetric complications and neonatal outcomes were compared among patients with different EMTs, which were measured the day before embryo thawing. The women were divided into three groups based on the EMT: group 1: < 9 mm; group 2: 9-12 mm; group 3: > 12 mm. Multiple logistic regression and subgroup analyses were performed to determine the potential confounding factors. RESULTS: The incidence of placenta previa in groups 1, 2, and 3 was 3.8, 1.0 and 0.5%, respectively, and that of cesarean section was 87.0, 78.3 and 72.0%, respectively (both P < 0.001). The gestational age and birth weight increased from group 1 to group 3 (both P < 0.001). After adjusting for confounders, a thicker EMT was found to be associated with a decreased risk of placenta previa (adjusted odds ratio (aOR) 0.798; 95% confidence interval (95% CI) 0.651-0.979; P = 0.031) and with a decreased risk of cesarean section (aOR 0.926; 95% CI 0.889-0.965; P < 0.001). Regarding the incidence of placenta previa, compared to women in group 3, women in group 1 had an aOR of 6.208 (95% CI 2.169-17.766; P = 0.001), and women in group 2 had an aOR of 1.862 (95% CI 0.851-4.076; P = 0.120). Regarding the incidence of cesarean section, compared to women in group 3, women in group 1 had an aOR of 2.111 (95% CI 1.415-3.455; P < 0.001), and women in group 2 had an aOR of 1.293 (95% CI 1.128-1.481; P < 0.001). Subgroup analyses showed similar results. CONCLUSIONS: Our results demonstrate that a thin endometrial lining is associated with adverse obstetric and neonatal outcomes and might be related to poor placentation.


Subject(s)
Embryo Transfer/methods , Endometrium/pathology , Placenta Previa/etiology , Adult , Cesarean Section , China , Female , Humans , Pregnancy , Retrospective Studies , Risk Factors
20.
Hum Reprod ; 33(5): 869-876, 2018 05 01.
Article in English | MEDLINE | ID: mdl-29546361

ABSTRACT

STUDY QUESTION: Does single cleavage-stage (Day 3) embryo transfer using a time-lapse (TL) hierarchical classification model achieve comparable ongoing pregnancy rates (OPR) to single blastocyst (Day 5) transfer by conventional morphological (CM) selection? SUMMARY ANSWER: Day 3 single embryo transfer (SET) with a hierarchical classification model had a significantly lower OPR compared with Day 5 SET with CM selection. WHAT IS KNOWN ALREADY: Cleavage-stage SET is an alternative to blastocyst SET. Time-lapse imaging assists better embryo selection, based on studies of pregnancy outcomes when adding time-lapse imaging to CM selection at the cleavage or blastocyst stage. STUDY DESIGN, SIZE, DURATION: This single-centre, randomized, open-label, active-controlled, non-inferiority study included 600 women between October 2015 and April 2017. PARTICIPANTS/MATERIALS, SETTING, METHODS: Eligible patients were Chinese females, aged ≤36 years, who were undergoing their first or second fresh IVF cycle using their own oocytes, and who had FSH levels ≤12 IU/mL on Day 3 of the cycle and 10 or more oocytes retrieved. Patients who had underlying uterine conditions, oocyte donation, recurrent pregnancy loss, abnormal oocytes or <6 normally fertilized embryos (2PN) were excluded from the study participation. Patients were randomized 1:1 to either the cleavage-stage SET with a time-lapse hierarchical classification model for selection (D3 + TL) or blastocyst SET with CM selection (D5 + CM). All normally fertilized zygotes were cultured in Primo Vision. The study was conducted at a tertiary IVF centre (CITIC-Xiangya) and OPR was the primary outcome. MAIN RESULTS AND THE ROLE OF CHANCE: A total of 600 patients were randomized to the two groups, among which 585 (D3 + TL = 290, D5 + CM = 295) were included in the Modified-intention-to-treat (mITT) population and 517 (D3 + TL = 261, D5 + CM = 256) were included in the PP population. In the per protocol (PP) population, OPR was significantly lower in the D3 group (59.4%, 155/261) than in the D5 group (68.4%, 175/256) (difference: -9.0%, 95% CI: -17.1%, -0.7%, P = 0.03). Analysis in mITT population showed a marginally significant difference in the OPR between the D3 + TL and D5 + CM groups (56.6 versus 64.1%, difference: -7.5%, 95% CI: -15.4%, 0.4%, P = 0.06). The D3 + TL group resulted in a markedly lower implantation rate than the D5 + CM group (64.4 versus 77.0%; P = 0.002) in the PP analysis, however, the early miscarriage rate did not significantly differ between the two groups. LIMITATIONS, REASONS FOR CAUTION: The study lacked a direct comparison between time-lapse and CM selections at cleavage-stage SET and was statistically underpowered to detect non-inferiority. The subject's eligibility criteria favouring women with a good prognosis for IVF weakened the generalizability of the results. WIDER IMPLICATIONS OF THE FINDINGS: The OPR from Day 3 cleavage-stage SET using hierarchical classification time-lapse selection was significantly lower compared with that from Day 5 blastocyst SET using conventional morphology, yet it appeared to be clinically acceptable in women underwent IVF. STUDY FUNDING/COMPETING INTEREST(S): This study is supported by grants from Ferring Pharmaceuticals and the Program for New Century Excellent Talents in University, China. TRIAL REGISTRATION NUMBER: ChiCTR-ICR-15006600. TRIAL REGISTRATION DATE: 16 June 2015. DATE OF FIRST PATIENT'S ENROLMENT: 1 October 2015.


Subject(s)
Embryo Implantation/physiology , Live Birth , Pregnancy Rate , Single Embryo Transfer/methods , Adult , Embryo Culture Techniques , Female , Humans , Pregnancy , Pregnancy Outcome , Time Factors , Time-Lapse Imaging , Treatment Outcome , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL