Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 35
Filter
1.
J Assist Reprod Genet ; 41(8): 2011-2020, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38951359

ABSTRACT

PURPOSE: Oocytes from women presenting primary ovarian insufficiency (POI) generate viable embryos at a lower rate than non-POI women, but the mechanisms responsible for the lower oocyte quality remain elusive. Due to the scarcity of human oocytes for research, animal models provide a promising way forward. We aimed at investigating the molecular events characterizing final maturation in POI oocytes in a well-defined POI-like bovine model. METHODS: Single-cell RNA-sequencing of bovine control and POI-like, GV, and MII oocytes (n = 5 per group) was performed. DEseq2 was used to identify differentially expressed genes. Further, a Gene set enrichment analysis and a transcriptomic meta-analysis between bovine and human oocytes were performed. RESULTS: In control cows, we found 2223 differentially expressed genes between the GV and MII stages. Specifically, the affected genes were related to RNA processing and transport, protein synthesis, organelle remodeling and reorganization, and metabolism. The meta-analysis with a set of young human oocytes at different maturation stages revealed 315 conserved genes through the GV-MII transition in cows and humans, mostly related to meiotic progression and cell cycle. Gene expression analysis between GV and MII of POI-like oocytes showed no differences in terms of differentially expressed genes, pointing towards a substantial failure to properly remodel the transcriptome in the POI model, and with the clustering analysis indicating that the cow's genetic background had a higher impact than the oocyte's maturation stage. CONCLUSION: Overall, we have identified and characterized a valuable animal model of POI, paving the way to identifying new molecular mechanisms involved in POI.


Subject(s)
Meiosis , Oocytes , Primary Ovarian Insufficiency , Cattle , Female , Primary Ovarian Insufficiency/genetics , Primary Ovarian Insufficiency/pathology , Animals , Oocytes/growth & development , Oocytes/metabolism , Oocytes/pathology , Meiosis/genetics , Humans , Transcriptome/genetics , Disease Models, Animal , Oogenesis/genetics
2.
Nutrients ; 16(11)2024 May 30.
Article in English | MEDLINE | ID: mdl-38892639

ABSTRACT

Compared to the general population, patients with inflammatory bowel disease (IBD) are less likely to be vaccinated, putting them at an increased risk of vaccine-preventable illnesses. This risk is further compounded by the immunosuppressive therapies commonly used in IBD management. Therefore, developing new treatments for IBD that maintain immune function is crucial, as successful management can lead to better vaccination outcomes and overall health for these patients. Here, we investigate the potential of recombinant banana lectin (rBanLec) as a supporting therapeutic measure to improve IBD control and possibly increase vaccination rates among IBD patients. By examining the therapeutic efficacy of rBanLec in a murine model of experimental colitis, we aim to lay the foundation for its application in improving vaccination outcomes. After inducing experimental colitis in C57BL/6 and BALB/c mice with 2,4,6-trinitrobenzene sulfonic acid, we treated animals orally with varying doses of rBanLec 0.1-10 µg/mL (0.01-1 µg/dose) during the course of the disease. We assessed the severity of colitis and rBanLec's modulation of the immune response compared to control groups. rBanLec administration resulted in an inverse dose-response reduction in colitis severity (less pronounced weight loss, less shortening of the colon) and an improved recovery profile, highlighting its therapeutic potential. Notably, rBanLec-treated mice exhibited significant modulation of the immune response, favoring anti-inflammatory pathways (primarily reduction in a local [TNFα]/[IL-10]) crucial for effective vaccination. Our findings suggest that rBanLec could mitigate the adverse effects of immunosuppressive therapy on vaccine responsiveness in IBD patients. By improving the underlying immune response, rBanLec may increase the efficacy of vaccinations, offering a dual benefit of disease management and prevention of vaccine-preventable illnesses. Further studies are required to translate these findings into clinical practice.


Subject(s)
Colitis , Disease Models, Animal , Inflammatory Bowel Diseases , Mice, Inbred BALB C , Mice, Inbred C57BL , Musa , Animals , Inflammatory Bowel Diseases/drug therapy , Mice , Musa/chemistry , Colitis/drug therapy , Colitis/immunology , Colitis/prevention & control , Plant Lectins/pharmacology , Trinitrobenzenesulfonic Acid , Immunomodulating Agents/pharmacology , Female , Colon/drug effects , Colon/immunology , Colon/pathology , Male
3.
Mol Hum Reprod ; 30(7)2024 Jun 26.
Article in English | MEDLINE | ID: mdl-38870523

ABSTRACT

Advanced maternal age is associated with a decline in oocyte quality, which often leads to reproductive failure in humans. However, the mechanisms behind this age-related decline remain unclear. To gain insights into this phenomenon, we applied plexDIA, a multiplexed data-independent acquisition, single-cell mass spectrometry method, to analyze the proteome of oocytes from both young women and women of advanced maternal age. Our findings primarily revealed distinct proteomic profiles between immature fully grown germinal vesicle and mature metaphase II oocytes. Importantly, we further show that a woman's age is associated with changes in her oocyte proteome. Specifically, when compared to oocytes obtained from young women, advanced maternal age oocytes exhibited lower levels of the proteasome and TRiC complex, as well as other key regulators of proteostasis and meiosis. This suggests that aging adversely affects the proteostasis and meiosis networks in human oocytes. The proteins identified in this study hold potential as targets for improving oocyte quality and may guide future studies into the molecular processes underlying oocyte aging.


Subject(s)
Maternal Age , Meiosis , Oocytes , Proteome , Proteomics , Proteostasis , Single-Cell Analysis , Humans , Oocytes/metabolism , Oocytes/cytology , Female , Meiosis/physiology , Adult , Proteomics/methods , Single-Cell Analysis/methods , Proteome/metabolism , Proteasome Endopeptidase Complex/metabolism , Middle Aged
4.
Trials ; 25(1): 38, 2024 Jan 11.
Article in English | MEDLINE | ID: mdl-38212837

ABSTRACT

BACKGROUND: Adequately selecting the initial follicle-stimulating hormone (FSH) dose during controlled ovarian stimulation (COS) is key for success in assisted reproduction. The objective of COS is to obtain an optimal number of oocytes to increase the chances of achieving a pregnancy, while avoiding complications for the patient. Current clinical protocols do achieve good results for the majority of patients, but further refinements in individualized FSH dosing may reduce the risk of poor ovarian response while also limiting the risk of ovarian hyperstimulation syndrome (OHSS) risk. Models to select the first FSH dose in COS have been presented in literature with promising results. However, most have only been developed and tested in normo-ovulatory women under the age of 40 years. METHODS: This is a randomized, controlled, multicenter, single blinded, clinical trial. This study will be performed in 236 first cycle in vitro fertilization (IVF) and/or ICSI (intracytoplasmic sperm injection) patients, randomized 1:1 in two arms. In the intervention arm, the dose of FSH will be assigned by a machine learning (ML) model called IDoser, while in the control arm, the dose will be determined by the clinician following standard practice. Stratified block randomization will be carried out depending on the patient being classified as expected low responder, high responder, or normo-responder. Patients will complete their participation in the trial once the first embryo transfer result is known. The primary outcome of the study is the number of metaphase II (MII) oocytes retrieved at ovarian pick up (OPU) and the hypothesis of non-inferiority of the intervention arm compared to the control. Secondary outcomes include the number of cycle cancelations (due to low response or no retrieval of mature oocytes), risk of ovarian hyperstimulation syndrome (OHSS), and clinical pregnancy and live birth rates per first transfer. DISCUSSION: To our knowledge, this is the first randomized trial to test clinical performance of an all-patient inclusive model to select the first dose of FSH for COS. Prospective trials for machine learning (ML) models in healthcare are scarce but necessary for clinical application. TRIAL REGISTRATION: ClinicalTrials.gov, NCT05948293 . Registered on 14 July 2023.


Subject(s)
Follicle Stimulating Hormone , Ovarian Hyperstimulation Syndrome , Male , Pregnancy , Humans , Female , Adult , Follicle Stimulating Hormone/adverse effects , Sperm Injections, Intracytoplasmic/methods , Ovarian Hyperstimulation Syndrome/etiology , Ovarian Hyperstimulation Syndrome/prevention & control , Prospective Studies , Ovulation Induction/adverse effects , Ovulation Induction/methods , Semen , Fertilization in Vitro/methods , Pregnancy Rate , Randomized Controlled Trials as Topic , Multicenter Studies as Topic
5.
Eur J Obstet Gynecol Reprod Biol ; 294: 163-169, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38266482

ABSTRACT

OBJECTIVE: Are circulating luteinizing hormone (LH) levels predictive of ovarian response in oocyte donors triggered with gonadotropin-releasing hormone (GnRH) agonists? STUDY DESIGN: A prospective cohort study with 224 oocyte donation cycles between 2021 and 2022 at a single center, examined the relationship between circulating luteinizing hormone (LH) levels and ovarian response. Oocyte donors underwent GnRH antagonist downregulation followed by GnRH agonist trigger. LH, estradiol, and progesterone levels were measured on day one of stimulation, trigger-day and 12 h post-trigger. Oocyte retrieval and maturity rates were analyzed using univariate and multivariate analyses, and the correlation between post-trigger LH levels and outcomes was assessed by Pearson's correlation test. A significance level of p < 0.05 was used. RESULTS: Mean age was 26 ± 4.3 years, mean body mass index (BMI, kg/m2) was 22.6 ± 3.2 and mean antral follicle count (AFC) was 21.7 ± 8.2. Post-trigger LH levels averaged 51.3 IU/L (SD 34.8), and oocyte retrieval rate and maturity rates were 112,7% (+/-48,1%) and 77,8% (+/- 17,2%), respectively. No significant differences were found in these outcomes for donors with post-trigger LH values below and above 15 IU/L (Mann Whitney's p > 0.05). However, exploratory analyses revealed that post-trigger LH values < 22 IU/L and basal LH levels < 4 IU/L were associated with significantly lower oocyte retrieval rate (90 % vs 110 %, p = 0.019 and 100 % vs 110 %, p = 0.019, respectively). CONCLUSIONS: This study, a first in exclusively focusing on oocyte donors, did not support the previously reported LH value of 15 IU/L as predictive of suboptimal ovarian response. TRIAL REGISTRATION NUMBER: ClinicalTrials.gov Identifier: NCT05109403.


Subject(s)
Gonadotropin-Releasing Hormone , Ovulation Induction , Female , Humans , Young Adult , Adult , Prospective Studies , Oocytes , Luteinizing Hormone , Fertilization in Vitro
6.
Hum Reprod ; 39(1): 258-274, 2024 Jan 05.
Article in English | MEDLINE | ID: mdl-37873575

ABSTRACT

STUDY QUESTION: Does the diagnosis of mosaicism affect ploidy rates across different providers offering preimplantation genetic testing for aneuploidies (PGT-A)? SUMMARY ANSWER: Our analysis of 36 395 blastocyst biopsies across eight genetic testing laboratories revealed that euploidy rates were significantly higher in providers reporting low rates of mosaicism. WHAT IS KNOWN ALREADY: Diagnoses consistent with chromosomal mosaicism have emerged as a third category of possible embryo ploidy outcomes following PGT-A. However, in the era of mosaicism, embryo selection has become increasingly complex. Biological, technical, analytical, and clinical complexities in interpreting such results have led to substantial variability in mosaicism rates across PGT-A providers and clinics. Critically, it remains unknown whether these differences impact the number of euploid embryos available for transfer. Ultimately, this may significantly affect clinical outcomes, with important implications for PGT-A patients. STUDY DESIGN, SIZE, DURATION: In this international, multicenter cohort study, we reviewed 36 395 consecutive PGT-A results, obtained from 10 035 patients across 11 867 treatment cycles, conducted between October 2015 and October 2021. A total of 17 IVF centers, across eight PGT-A providers, five countries and three continents participated in the study. All blastocysts were tested using trophectoderm biopsy and next-generation sequencing. Both autologous and donation cycles were assessed. Cycles using preimplantation genetic testing for structural rearrangements were excluded from the analysis. PARTICIPANTS/MATERIALS, SETTING, METHODS: The PGT-A providers were randomly categorized (A to H). Providers B, C, D, E, F, G, and H all reported mosaicism, whereas Provider A reported embryos as either euploid or aneuploid. Ploidy rates were analyzed using multilevel mixed linear regression. Analyses were adjusted for maternal age, paternal age, oocyte source, number of embryos biopsied, day of biopsy, and PGT-A provider, as appropriate. We compared associations between genetic testing providers and PGT-A outcomes, including the number of chromosomally normal (euploid) embryos determined to be suitable for transfer. MAIN RESULTS AND THE ROLE OF CHANCE: The mean maternal age (±SD) across all providers was 36.2 (±5.2). Our findings reveal a strong association between PGT-A provider and the diagnosis of euploidy and mosaicism. Amongst the seven providers that reported mosaicism, the rates varied from 3.1% to 25.0%. After adjusting for confounders, we observed a significant difference in the likelihood of diagnosing mosaicism across providers (P < 0.001), ranging from 6.5% (95% CI: 5.2-7.4%) for Provider B to 35.6% (95% CI: 32.6-38.7%) for Provider E. Notably, adjusted euploidy rates were highest for providers that reported the lowest rates of mosaicism (Provider B: euploidy, 55.7% (95% CI: 54.1-57.4%), mosaicism, 6.5% (95% CI: 5.2-7.4%); Provider H: euploidy, 44.5% (95% CI: 43.6-45.4%), mosaicism, 9.9% (95% CI: 9.2-10.6%)); and Provider D: euploidy, 43.8% (95% CI: 39.2-48.4%), mosaicism, 11.0% (95% CI: 7.5-14.5%)). Moreover, the overall chance of having at least one euploid blastocyst available for transfer was significantly higher when mosaicism was not reported, when we compared Provider A to all other providers (OR = 1.30, 95% CI: 1.13-1.50). Differences in diagnosing and interpreting mosaic results across PGT-A laboratories raise further concerns regarding the accuracy and relevance of mosaicism predictions. While we confirmed equivalent clinical outcomes following the transfer of mosaic and euploid blastocysts, we found that a significant proportion of mosaic embryos are not used for IVF treatment. LIMITATIONS, REASONS FOR CAUTION: Due to the retrospective nature of the study, associations can be ascertained, however, causality cannot be established. Certain parameters such as blastocyst grade were not available in the dataset. Furthermore, certain platform-related and clinic-specific factors may not be readily quantifiable or explicitly captured in our dataset. As such, a full elucidation of all potential confounders accounting for variability may not be possible. WIDER IMPLICATIONS OF THE FINDINGS: Our findings highlight the strong need for standardization and quality assurance in the industry. The decision not to transfer mosaic embryos may ultimately reduce the chance of success of a PGT-A cycle by limiting the pool of available embryos. Until we can be certain that mosaic diagnoses accurately reflect biological variability, reporting mosaicism warrants utmost caution. A prudent approach is imperative, as it may determine the difference between success or failure for some patients. STUDY FUNDING/COMPETING INTEREST(S): This work was supported by the Torres Quevedo Grant, awarded to M.P. (PTQ2019-010494) by the Spanish State Research Agency, Ministry of Science and Innovation, Spain. M.P., L.B., A.R.L., A.L.R.d.C.L., N.P.P., M.P., D.S., F.A., A.P., B.M., L.D., F.V.M., D.S., M.R., E.P.d.l.B., A.R., and R.V. have no competing interests to declare. B.L., R.M., and J.A.O. are full time employees of IB Biotech, the genetics company of the Instituto Bernabeu group, which performs preimplantation genetic testing. M.G. is a full time employee of Novagen, the genetics company of Cegyr, which performs preimplantation genetic testing. TRIAL REGISTRATION NUMBER: N/A.


Subject(s)
Mosaicism , Preimplantation Diagnosis , Female , Humans , Pregnancy , Aneuploidy , Bias, Implicit , Blastocyst/pathology , Cohort Studies , Genetic Testing/methods , Preimplantation Diagnosis/methods , Retrospective Studies , Adult
8.
Mol Ther ; 31(8): 2326-2341, 2023 08 02.
Article in English | MEDLINE | ID: mdl-37376733

ABSTRACT

Human germline gene correction by targeted nucleases holds great promise for reducing mutation transmission. However, recent studies have reported concerning observations in CRISPR-Cas9-targeted human embryos, including mosaicism and loss of heterozygosity (LOH). The latter has been associated with either gene conversion or (partial) chromosome loss events. In this study, we aimed to correct a heterozygous basepair substitution in PLCZ1, related to infertility. In 36% of the targeted embryos that originated from mutant sperm, only wild-type alleles were observed. By performing genome-wide double-digest restriction site-associated DNA sequencing, integrity of the targeted chromosome (i.e., no deletions larger than 3 Mb or chromosome loss) was confirmed in all seven targeted GENType-analyzed embryos (mutant editing and absence of mutation), while short-range LOH events (shorter than 10 Mb) were clearly observed by single-nucleotide polymorphism assessment in two of these embryos. These results fuel the currently ongoing discussion on double-strand break repair in early human embryos, making a case for the occurrence of gene conversion events or partial template-based homology-directed repair.


Subject(s)
CRISPR-Cas Systems , Gene Editing , Humans , Male , Gene Editing/methods , Semen , Mutation , Alleles , Chromosomes
9.
Mol Hum Reprod ; 29(7)2023 Jun 30.
Article in English | MEDLINE | ID: mdl-37261882

ABSTRACT

Human meiosis in oocytes entails an intricate regulation of the transcriptome to support late oocyte growth and early embryo development, both crucial to reproductive success. Currently, little is known about the co- and post-transcriptional mRNA processing mechanisms regulating the last meiotic phases, which contribute to transcriptome complexity and influence translation rates. We analyzed gene expression changes, splicing and pre-mRNA processing in an RNA sequencing set of 40 human oocytes at different meiotic maturation stages, matured both in vivo and in vitro. We found abundant untranslated region (UTR) processing, mostly at the 3' end, of meiosis-related genes between the germinal vesicle (GV) and metaphase II (MII) stages, supported by the differential expression of spliceosome and pre-mRNA processing related genes. Importantly, we found very few differences among GV oocytes across several durations of IVM, as long as they did not reach MII, suggesting an association of RNA processing and successful meiosis transit. Changes in protein isoforms are minor, although specific and consistent for genes involved in chromosome organization and spindle assembly. In conclusion, we reveal a dynamic transcript remodeling during human female meiosis, and show how pre-mRNA processing, specifically 3'UTR shortening, drives a selective translational regulation of transcripts necessary to reach final meiotic maturation.


Subject(s)
In Vitro Oocyte Maturation Techniques , RNA Precursors , Humans , Female , RNA Precursors/genetics , RNA Precursors/metabolism , Oocytes/metabolism , Meiosis/genetics , Oogenesis/genetics
11.
Placenta ; 125: 36-46, 2022 07.
Article in English | MEDLINE | ID: mdl-35078656

ABSTRACT

Human reproductive success relies on the intricate interplay between the developing embryo and the maternal endometrium. These highly-coordinated interactions facilitate implantation, setting in motion a series of developmental programs to establish a sustained fetal-maternal interface. Understanding endometrial function and the early human embryo-maternal dialogue is thus an important prerequisite for refining clinical approaches to alleviate implantation failure, early pregnancy loss and other obstetric complications. Yet, many mediators of implantation remain elusive. Driven by endocrine factors, interactions at the embryo-maternal interface are tightly regulated and highly complex. Coupled to the inaccessibility of the in vivo environment and scarcity of research material, studying human implantation remains exceptionally challenging. Nevertheless, the field continues to gain momentum. Cutting-edge omics technologies and high-resolution imaging have revealed important structural and functional insights into endometrial biology, while emerging bioengineering tools are enhancing our ability to model the synergies and individual features of the embryo-maternal environment. Novel in vitro platforms using human cells and embryos are considerably more accessible and easier to manipulate compared to in vivo approaches, enhancing our ability to capture specific stages of implantation. This review aims to showcase current and emerging technologies used to study human endometrial biology and the early embryo-maternal interface, including single cell omics approaches, bioengineered endometrial models and embryo-endometrium co-culture platforms. We highlight the value of these approaches and provide our perspective on the current challenges faced by the field. Recognizing the physiological scope of these emerging technologies will be key for utilizing their full potential and driving future innovation.


Subject(s)
Abortion, Spontaneous , Endometrium , Coculture Techniques , Embryo Implantation/physiology , Embryo, Mammalian , Female , Humans , Pregnancy
12.
J Assist Reprod Genet ; 39(3): 609-618, 2022 Mar.
Article in English | MEDLINE | ID: mdl-35064435

ABSTRACT

PURPOSE: Providing additional insights on the efficacy of human nuclear transfer (NT). Here, and earlier, NT has been applied to minimize transmission risk of mitochondrial DNA (mtDNA) diseases. NT has also been proposed for treating infertility, but it is still unclear which infertility indications would benefit. In this work, we therefore additionally assess the applicability of NT to overcome failed fertilization. METHODS: Patient 1 carries a homoplasmic mtDNA mutation (m.11778G > A). Seventeen metaphase II (MII) oocytes underwent pre-implantation genetic testing (PGT), while five MII oocytes were used for spindle transfer (ST), and one in vitro matured (IVM) metaphase I oocyte underwent early pronuclear transfer (ePNT). Patients 2-3 experienced multiple failed intracytoplasmic sperm injection (ICSI) and ICSI-assisted oocyte activation (AOA) cycles. For these patients, the obtained MII oocytes underwent an additional ICSI-AOA cycle, while the IVM oocytes were subjected to ST. RESULTS: For patient 1, PGT-M confirmed mutation loads close to 100%. All ST-reconstructed oocytes fertilized and cleaved, of which one progressed to the blastocyst stage. The reconstructed ePNT-zygote reached the morula stage. These samples showed an average mtDNA carry-over rate of 2.9% ± 0.8%, confirming the feasibility of NT to reduce mtDNA transmission. For patient 2-3 displaying fertilization failure, ST resulted in, respectively, 4/5 and 6/6 fertilized oocytes, providing evidence, for the first time, that NT can enable successful fertilization in this patient population. CONCLUSION: Our study showcases the repertoire of disorders for which NT can be beneficial, to overcome either mitochondrial disease transmission or failed fertilization after ICSI-AOA.


Subject(s)
Infertility , Mitochondrial Diseases , DNA, Mitochondrial/genetics , Fertilization , Fertilization in Vitro/methods , Humans , Infertility/genetics , Infertility/therapy , Oocytes , Sperm Injections, Intracytoplasmic
15.
Int J Biol Macromol ; 183: 423-434, 2021 Jul 31.
Article in English | MEDLINE | ID: mdl-33932415

ABSTRACT

In this study, Lactobacillus reuteri B2 was isolated from the feces of C57BL/6 mice and assessed on probiotic activity. L. reuteri B2 was identified by 16S rDNA sequencing, which the cell viability in acidic conditions at pH 2.0 was 64% after 2 h, and in the presents of 0.30% of the bile salts, after 6 h, was 37%. Antimicrobial assay with L. reuteri B2 showed maximum diameters against Klebsiela oxytoca J7 (12.5 ± 0.71 mm). We further hypothesized if L. reuteri B2 strain in the free form can survive all conditions in the gastrointestinal tract (GIT) then the utilization of the appropriate biomaterials would ameliorate its stability and viability in GIT. L. reuteri B2 was microencapsulated into sodium alginate-(Na-alg) and different content of Na-alg and sodium maleate (SM) beads. Characterization materials enveloped their thermal characteristics (TGA/DTA analysis) and structure using: scanning electron microscopy (SEM), FTIR, and particle size distribution. The high survival rate of L. reuteri B2 at low pH from 2.0 to 4.0 and in the presence of the bile salts, at concentrations up to 0.30%, was obtained. L. reuteri B2 showed strong antimicrobial activity and the best protection microencapsulated with Na-alg + SM in simulated gastric juices (SGJ).


Subject(s)
Alginates/chemistry , Biopolymers/chemistry , DNA, Ribosomal/metabolism , Limosilactobacillus reuteri/metabolism , Probiotics/therapeutic use
16.
Nat Commun ; 12(1): 1286, 2021 02 24.
Article in English | MEDLINE | ID: mdl-33627650

ABSTRACT

DNA methylation (5mC) is central to cellular identity. The global erasure of 5mC from the parental genomes during preimplantation mammalian development is critical to reset the methylome of gametes to the cells in the blastocyst. While active and passive modes of demethylation have both been suggested to play a role in this process, the relative contribution of these two mechanisms to 5mC erasure remains unclear. Here, we report a single-cell method (scMspJI-seq) that enables strand-specific quantification of 5mC, allowing us to systematically probe the dynamics of global demethylation. When applied to mouse embryonic stem cells, we identified substantial cell-to-cell strand-specific 5mC heterogeneity, with a small group of cells displaying asymmetric levels of 5mCpG between the two DNA strands of a chromosome suggesting loss of maintenance methylation. Next, in preimplantation mouse embryos, we discovered that methylation maintenance is active till the 16-cell stage followed by passive demethylation in a fraction of cells within the early blastocyst at the 32-cell stage of development. Finally, human preimplantation embryos qualitatively show temporally delayed yet similar demethylation dynamics as mouse embryos. Collectively, these results demonstrate that scMspJI-seq is a sensitive and cost-effective method to map the strand-specific genome-wide patterns of 5mC in single cells.


Subject(s)
DNA Demethylation , DNA Methylation/physiology , Animals , Blastocyst/metabolism , DNA (Cytosine-5-)-Methyltransferase 1/deficiency , DNA (Cytosine-5-)-Methyltransferase 1/genetics , DNA Methylation/genetics , Embryonic Development/genetics , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Female , Humans , Mice , Mice, Knockout , Pregnancy
17.
Stem Cells ; 39(5): 551-563, 2021 05.
Article in English | MEDLINE | ID: mdl-33470497

ABSTRACT

Protocols for specifying human primordial germ cell-like cells (hPGCLCs) from human embryonic stem cells (hESCs) remain hindered by differences between hESC lines, their derivation methods, and maintenance culture conditions. This poses significant challenges for establishing reproducible in vitro models of human gametogenesis. Here, we investigated the influence of activin A (ActA) during derivation and maintenance on the propensity of hESCs to differentiate into PGCLCs. We show that continuous ActA supplementation during hESC derivation (from blastocyst until the formation of the post-inner cell mass intermediate [PICMI]) and supplementation (from the first passage of the PICMI onwards) is beneficial to differentiate hESCs to PGCLCs subsequently. Moreover, comparing isogenic primed and naïve states prior to differentiation, we showed that conversion of hESCs to the 4i-state improves differentiation to (TNAP [tissue nonspecific alkaline phosphatase]+/PDPN [podoplanin]+) PGCLCs. Those PGCLCs expressed several germ cell markers, including TFAP2C (transcription factor AP-2 gamma), SOX17 (SRY-box transcription factor 17), and NANOS3 (nanos C2HC-type zinc finger 3), and markers associated with germ cell migration, CXCR4 (C-X-C motif chemokine receptor 4), LAMA4 (laminin subunit alpha 4), ITGA6 (integrin subunit alpha 6), and CDH4 (cadherin 4), suggesting that the large numbers of PGCLCs obtained may be suitable to differentiate further into more mature germ cells. Finally, hESCs derived in the presence of ActA showed higher competence to differentiate to hPGCLC, in particular if transiently converted to the 4i-state. Our work provides insights into the differences in differentiation propensity of hESCs and delivers an optimized protocol to support efficient human germ cell derivation.


Subject(s)
Activins/genetics , Cell Differentiation/genetics , Germ Cells/cytology , Human Embryonic Stem Cells/cytology , Blastocyst/cytology , Cadherins/genetics , Cells, Cultured , Gene Expression Regulation, Developmental/genetics , Germ Cells/growth & development , Human Embryonic Stem Cells/metabolism , Humans , Integrin alpha6/genetics , Laminin/genetics , RNA-Binding Proteins/genetics , Receptors, CXCR4/genetics , SOXF Transcription Factors/genetics , Signal Transduction/genetics , Transcription Factor AP-2/genetics
18.
Genes Chromosomes Cancer ; 60(4): 272-281, 2021 04.
Article in English | MEDLINE | ID: mdl-33336840

ABSTRACT

Human embryonic stem cells (hESCs) and embryonal tumors share a number of common features, including a compromised G1/S checkpoint. Consequently, these rapidly dividing hESCs and cancer cells undergo elevated levels of replicative stress, inducing genomic instability that drives chromosomal imbalances. In this context, it is of interest that long-term in vitro cultured hESCs exhibit a remarkable high incidence of segmental DNA copy number gains, some of which are also highly recurrent in certain malignancies such as 17q gain (17q+). The selective advantage of DNA copy number changes in these cells has been attributed to several underlying processes including enhanced proliferation. We hypothesized that these recurrent chromosomal imbalances become rapidly embedded in the cultured hESCs through a replicative stress driven Darwinian selection process. To this end, we compared the effect of hydroxyurea-induced replicative stress vs normal growth conditions in an equally mixed cell population of isogenic euploid and 17q + hESCs. We could show that 17q + hESCs rapidly overtook normal hESCs. Our data suggest that recurrent chromosomal segmental gains provide a proliferative advantage to hESCs under increased replicative stress, a process that may also explain the highly recurrent nature of certain imbalances in cancer.


Subject(s)
Cell Division , Chromosome Aberrations , Human Embryonic Stem Cells/cytology , Selection, Genetic , Cell Cycle/genetics , Cell Line, Tumor , Cell Proliferation , Chromosomes, Human, Pair 17 , DNA Copy Number Variations , Humans , Hydroxyurea , Stress, Physiological , Transcriptome
19.
Hum Reprod Update ; 26(6): 779-798, 2020 11 01.
Article in English | MEDLINE | ID: mdl-32712668

ABSTRACT

BACKGROUND: Studying the human peri-implantation period remains hindered by the limited accessibility of the in vivo environment and scarcity of research material. As such, continuing efforts have been directed towards developing embryo-like structures (ELS) from pluripotent stem cells (PSCs) that recapitulate aspects of embryogenesis in vitro. While the creation of such models offers immense potential for studying fundamental processes in both pre- and early post-implantation development, it also proves ethically contentious due to wide-ranging views on the moral and legal reverence due to human embryos. Lack of clarity on how to qualify and regulate research with ELS thus presents a challenge in that it may either limit this new field of research without valid grounds or allow it to develop without policies that reflect justified ethical concerns. OBJECTIVE AND RATIONALE: The aim of this article is to provide a comprehensive overview of the existing scientific approaches to generate ELS from mouse and human PSCs, as well as discuss future strategies towards innovation in the context of human development. Concurrently, we aim to set the agenda for the ethical and policy issues surrounding research on human ELS. SEARCH METHODS: The PubMed database was used to search peer-reviewed articles and reviews using the following terms: 'stem cells', 'pluripotency', 'implantation', 'preimplantation', 'post-implantation', 'blastocyst', 'embryoid bodies', 'synthetic embryos', 'embryo models', 'self-assembly', 'human embryo-like structures', 'artificial embryos' in combination with other keywords related to the subject area. The PubMed and Web of Science databases were also used to systematically search publications on the ethics of ELS and human embryo research by using the aforementioned keywords in combination with 'ethics', 'law', 'regulation' and equivalent terms. All relevant publications until December 2019 were critically evaluated and discussed. OUTCOMES: In vitro systems provide a promising way forward for uncovering early human development. Current platforms utilize PSCs in both two- and three-dimensional settings to mimic various early developmental stages, including epiblast, trophoblast and amniotic cavity formation, in addition to axis development and gastrulation. Nevertheless, much hinges on the term 'embryo-like'. Extension of traditional embryo frameworks to research with ELS reveals that (i) current embryo definitions require reconsideration, (ii) cellular convertibility challenges the attribution of moral standing on the basis of 'active potentiality' and (iii) meaningful application of embryo protective directives will require rethinking of the 14-day culture limit and moral weight attributed to (non-)viability. Many conceptual and normative (dis)similarities between ELS and embryos thus remain to be thoroughly elucidated. WIDER IMPLICATIONS: Modelling embryogenesis holds vast potential for both human developmental biology and understanding various etiologies associated with infertility. To date, ELS have been shown to recapitulate several aspects of peri-implantation development, but critically, cannot develop into a fetus. Yet, concurrent to scientific innovation, considering the extent to which the use of ELS may raise moral concerns typical of human embryo research remains paramount. This will be crucial for harnessing the potential of ELS as a valuable research tool, whilst remaining within a robust moral and legal framework of professionally acceptable practices.


Subject(s)
Embryo Research/ethics , Embryo, Mammalian/cytology , Embryonic Development/physiology , Models, Biological , Public Policy , Animals , Embryo Implantation/physiology , Embryo Research/legislation & jurisprudence , Humans , Mice , Morals
SELECTION OF CITATIONS
SEARCH DETAIL