Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Mais filtros

Base de dados
Ano de publicação
Tipo de documento
Intervalo de ano de publicação
1.
Brain ; 2024 Jun 04.
Artigo em Inglês | MEDLINE | ID: mdl-38833623

RESUMO

Congenital hydrocephalus (CH), characterized by cerebral ventriculomegaly, is one of the most common reasons for pediatric brain surgery. Recent studies have implicated lin-41 (lineage variant 41)/TRIM71 (tripartite motif 71) as a candidate CH risk gene, however, TRIM71 variants have not been systematically examined in a large patient cohort or conclusively linked with an OMIM syndrome. Through cross-sectional analysis of the largest assembled cohort of patients with cerebral ventriculomegaly, including neurosurgically-treated CH (totaling 2,697 parent-proband trios and 8,091 total exomes), we identified 13 protein-altering de novo variants (DNVs) in TRIM71 in unrelated children exhibiting variable ventriculomegaly, CH, developmental delay, dysmorphic features, and other structural brain defects including corpus callosum dysgenesis and white matter hypoplasia. Eight unrelated patients were found to harbor arginine variants, including two recurrent missense DNVs, at homologous positions in RPXGV motifs of different NHL domains. Seven additional patients with rare, damaging, unphased or transmitted variants of uncertain significance were also identified. NHL-domain variants of TRIM71 exhibited impaired binding to the canonical TRIM71 target CDKN1A; other variants failed to direct the subcellular localization of TRIM71 to processing bodies. Single-cell transcriptomic analysis of human embryos revealed expression of TRIM71 in early first-trimester neural stem cells of the brain. These data show TRIM71 is essential for human brain morphogenesis and that TRIM71 mutations cause a novel neurodevelopmental syndrome featuring ventriculomegaly and CH.

2.
Biol Reprod ; 110(5): 936-949, 2024 May 09.
Artigo em Inglês | MEDLINE | ID: mdl-38271627

RESUMO

The placenta, composed of chorionic villi, changes dramatically across gestation. Understanding differences in ongoing pregnancies are essential to identify the role of chorionic villi at specific times in gestation and develop biomarkers and prognostic indicators of maternal-fetal health. The normative mRNA profile is established using next-generation sequencing of 124 first trimester and 43 third trimester human placentas from ongoing healthy pregnancies. Stably expressed genes (SEGs) not different between trimesters and with low variability are identified. Differential expression analysis of first versus third trimester adjusted for fetal sex is performed, followed by a subanalysis with 23 matched pregnancies to control for subject variability using the same genetic and environmental background. Placenta expresses 14,979 polyadenylated genes above sequencing noise (transcripts per million > 0.66), with 10.7% SEGs across gestation. Differentially expressed genes (DEGs) account for 86.7% of genes in the full cohort [false discovery rate (FDR) < 0.05]. Fold changes highly correlate between the full cohort and subanalysis (Pearson = 0.98). At stricter thresholds (FDR < 0.001, fold change > 1.5), there remains 50.1% DEGs (3353 upregulated in first and 4155 upregulated in third trimester). This is the largest mRNA atlas of healthy human placenta across gestation, controlling for genetic and environmental factors, demonstrating substantial changes from first to third trimester in chorionic villi. Specific differences and SEGs may be used to understand the specific role of the chorionic villi throughout gestation and develop first trimester biomarkers of placental health that transpire across gestation, which can be used for future development of biomarkers for maternal-fetal health.


Assuntos
Placenta , Primeiro Trimestre da Gravidez , Terceiro Trimestre da Gravidez , RNA Mensageiro , Transcriptoma , Humanos , Feminino , Gravidez , Terceiro Trimestre da Gravidez/genética , Placenta/metabolismo , RNA Mensageiro/metabolismo , RNA Mensageiro/genética , Primeiro Trimestre da Gravidez/genética , Adulto , Sequenciamento de Nucleotídeos em Larga Escala
3.
Obstet Gynecol ; 2024 Aug 22.
Artigo em Inglês | MEDLINE | ID: mdl-39173182

RESUMO

OBJECTIVE: To compare pregnancy and neonatal outcomes in women with hyperandrogenic polycystic ovarian syndrome (PCOS) phenotypes compared with nonhyperandrogenic PCOS phenotypes. METHODS: We conducted a retrospective cohort study of participants in the PPCOS (Pregnancy in Polycystic Ovary Syndrome) I and II randomized controlled trials; all of the participants met the National Institutes of Health diagnostic criteria for PCOS and were then sorted into three of the four Rotterdam criteria categories based on medical interview, demographics, physical examination, and laboratory data. The two hyperandrogenic (A and B) Rotterdam categories were compared with the nonhyperandrogenic phenotype of PCOS (phenotype D). Our outcomes of interest were clinical pregnancy, pregnancy loss, live birth, obstetric complications (including preterm labor, preeclampsia, gestational diabetes, intrauterine growth restriction, and premature rupture of membranes), and neonatal outcomes (including jaundice, respiratory distress syndrome, neonatal hospitalization, and neonatal infection). RESULTS: Of the 1,376 participants included in the study, 1,249 (90.8%) had hyperandrogenic PCOS phenotypes compared with 127 (9.2%) nonhyperandrogenic PCOS (nonhyperandrogenic PCOS). Compared with participants with nonhyperandrogenic PCOS, those with hyperandrogenic PCOS had higher body mass index (BMI) (35.5±8.9 vs 31.9±9.3 kg/m2, P<.001), fasting insulin (21.6±27.7 vs 14.7±15.0 micro-international units/mL, P<.001), and homeostatic model assessment for insulin resistance score (5.01±9.1 vs 3.4±4.1, P=.0002). Age and race were similar between groups. Months attempting pregnancy were greater in participants with hyperandrogenic PCOS compared with nonhyperandrogenic PCOS (41.8±37.3 vs 33.9±32.0). The proportion of participants who achieved pregnancy (29.9% vs 40.2%, P=.02) and live birth rates (20.1% vs 33.1%, P=.001) were lower among those with hyperandrogenic PCOS compared with nonhyperandrogenic PCOS, although pregnancy loss rates did not differ significantly (23.9% vs 32.3%, P=.06). The hyperandrogenic PCOS group had lower odds of live birth compared with the nonhyperandrogenic PCOS group (odds ratio [OR] 0.51, CI, 0.34-0.76), even after adjusting for BMI (adjusted odds ratio [aOR] 0.59, CI, 0.40-0.89). The hyperandrogenic PCOS group also had lower odds of achieving pregnancy compared with the nonhyperandrogenic PCOS group (OR 0.63, CI, 0.44-0.92); however, this association was no longer significant after adjusting for BMI (aOR 0.74, CI, 0.50-1.10). The overall low prevalence of prenatal complications and neonatal outcomes precluded a meaningful comparison between the two groups. CONCLUSION: Participants with hyperandrogenic PCOS achieved lower rates of pregnancy and live birth compared with those with nonhyperandrogenic PCOS. Evaluating distinct PCOS phenotypes may allow for individualized guidance regarding the probability of pregnancy and live birth. CLINICAL TRIALS REGISTRATION: ClinicalTrials.gov, NCT00068861 and NCT00718186.

4.
Fertil Steril ; 121(6): 1040-1052, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38307453

RESUMO

OBJECTIVE: To determine whether alterations in nonesterified fatty acid (NEFA) dynamics or degree of hyperandrogenism (HA) contribute to the difference in insulin sensitivity between women with metabolically healthy obese polycystic ovary syndrome (PCOS) (MHO-PCOS) and women with metabolically unhealthy obese PCOS (MUO-PCOS). DESIGN: Prospective cross-sectional study. SETTING: Tertiary-care academic center. PATIENTS: One hundred twenty-five obese women with PCOS. INTERVENTION: Consecutive obese (body mass index [BMI] ≥ 30 kg/m2) oligo-ovulatory women (n = 125) with PCOS underwent an oral glucose tolerance test and a subgroup of 16 participants underwent a modified frequently sampled intravenous glucose tolerance test to determine insulin-glucose and -NEFA dynamics. MAIN OUTCOME MEASURES: Degree of insulin resistance (IR) in adipose tissue (AT) basally (Adipo-IR) and dynamically (the nadir in NEFA levels observed [NEFAnadir], the time it took for NEFA levels to reach nadir [TIMEnadir], and the percent suppression in plasma NEFA levels from baseline to nadir [%NEFAsupp]); peak lipolysis rate (SNEFA) and peak rate of NEFA disposal from plasma pool (KNEFA); whole-body insulin-glucose interaction (acute response of insulin to glucose [AIRg], insulin sensitivity index [Si], glucose effectiveness [Sg], and disposition index [Di]); and HA (hirsutism score, total and free testosterone levels, and dehydroepiandrosterone sulfate levels). RESULTS: A total of 85 (68%) women were MUO-PCOS and 40 (32%) were MHO-PCOS using the homeostasis model of assessment of IR. Subjects with MUO-PCOS and MHO-PCOS did not differ in mean age, BMI, waist-to-hip ratio, HA, and lipoprotein levels. By a modified frequently sampled intravenous glucose tolerance test, eight women with MUO-PCOS had lesser Si, KNEFA, and the percent suppression in plasma NEFA levels from baseline to nadir (%NEFAsupp) and greater TIMEnadir, NEFAnadir, and baseline adipose tissue IR index (Adipo-IR) than eight subjects with MHO-PCOS, but similar fasting NEFA levels and SNEFA. Women with MUO-PCOS had a higher homeostasis model of assessment-ß% and fasting insulin levels than women with MHO-PCOS. In bivalent analysis, Si correlated strongly and negatively with Adipo-IR and NEFAnadir, weakly and negatively with TIMEnadir, and positively with KNEFA and %NEFAsupp, in women with MUO-PCOS only. CONCLUSION: Independent of age and BMI, women with MUO-PCOS have reduced NEFA uptake and altered insulin-mediated NEFA suppression, but no difference in HA, compared with women with MHO-PCOS. Altered insulin-mediated NEFA suppression, rather than HA or lipolysis rate, contributes to variations in insulin sensitivity among obese women with PCOS.


Assuntos
Ácidos Graxos não Esterificados , Hiperandrogenismo , Resistência à Insulina , Obesidade , Síndrome do Ovário Policístico , Humanos , Feminino , Síndrome do Ovário Policístico/metabolismo , Síndrome do Ovário Policístico/sangue , Síndrome do Ovário Policístico/complicações , Hiperandrogenismo/metabolismo , Hiperandrogenismo/sangue , Adulto , Ácidos Graxos não Esterificados/sangue , Ácidos Graxos não Esterificados/metabolismo , Obesidade/metabolismo , Obesidade/sangue , Obesidade/complicações , Estudos Transversais , Resistência à Insulina/fisiologia , Estudos Prospectivos , Adulto Jovem , Teste de Tolerância a Glucose , Glicemia/metabolismo , Insulina/sangue , Biomarcadores/sangue
5.
medRxiv ; 2024 Jul 23.
Artigo em Inglês | MEDLINE | ID: mdl-39108523

RESUMO

Human trophoblast stem (TS) cells are an informative in vitro model for the generation and testing of biologically meaningful hypotheses. The goal of this project was to derive patient-specific TS cell lines from clinically available chorionic villus sampling biopsies. Cell outgrowths were captured from human chorionic villus tissue specimens cultured in modified human TS cell medium. Cell colonies emerged early during the culture and cell lines were established and passaged for several generations. Karyotypes of the newly established chorionic villus-derived trophoblast stem (TS CV ) cell lines were determined and compared to initial genetic diagnoses from freshly isolated chorionic villi. Phenotypes of TSCV cells in the stem state and following differentiation were compared to cytotrophoblast-derived TS (TS CT ) cells. TSCV and TSCT cells uniformly exhibited similarities in the stem state and following differentiation into syncytiotrophoblast and extravillous trophoblast cells. Chorionic villus tissue specimens provide a valuable source for TS cell derivation. They expand the genetic diversity of available TS cells and are associated with defined clinical outcomes. TSCV cell lines provide a new set of experimental tools for investigating trophoblast cell lineage development.

6.
Placenta ; 150: 8-21, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38537412

RESUMO

INTRODUCTION: Fetal sex affects fetal and maternal health outcomes in pregnancy, but this connection remains poorly understood. As the placenta is the route of fetomaternal communication and derives from the fetal genome, placental gene expression sex differences may explain these outcomes. OBJECTIVES: We utilized next generation sequencing to study the normal human placenta in both sexes in first and third trimester to generate a normative transcriptome based on sex and gestation. STUDY DESIGN: We analyzed 124 first trimester (T1, 59 female and 65 male) and 43 third trimester (T3, 18 female and 25 male) samples for sex differences within each trimester and sex-specific gestational differences. RESULTS: Placenta shows more significant sexual dimorphism in T1, with 94 T1 and 26 T3 differentially expressed genes (DEGs). The sex chromosomes contributed 60.6% of DEGs in T1 and 80.8% of DEGs in T3, excluding X/Y pseudoautosomal regions. There were 6 DEGs from the pseudoautosomal regions, only significant in T1 and all upregulated in males. The distribution of DEGs on the X chromosome suggests genes on Xp (the short arm) may be particularly important in placental sex differences. Dosage compensation analysis of X/Y homolog genes shows expression is primarily contributed by the X chromosome. In sex-specific analyses of first versus third trimester, there were 2815 DEGs common to both sexes upregulated in T1, and 3263 common DEGs upregulated in T3. There were 7 female-exclusive DEGs upregulated in T1, 15 female-exclusive DEGs upregulated in T3, 10 male-exclusive DEGs upregulated in T1, and 20 male-exclusive DEGs upregulated in T3. DISCUSSION: This is the largest cohort of placentas across gestation from healthy pregnancies defining the normative sex dimorphic gene expression and sex common, sex specific and sex exclusive gene expression across gestation. The first trimester has the most sexually dimorphic transcripts, and the majority were upregulated in females compared to males in both trimesters. The short arm of the X chromosome and the pseudoautosomal region is particularly critical in defining sex differences in the first trimester placenta. As pregnancy is a dynamic state, sex specific DEGs across gestation may contribute to sex dimorphic changes in overall outcomes.


Assuntos
Sequenciamento de Nucleotídeos em Larga Escala , Placenta , Caracteres Sexuais , Humanos , Feminino , Gravidez , Masculino , Placenta/metabolismo , RNA Mensageiro/metabolismo , RNA Mensageiro/genética , Adulto , Transcriptoma , Terceiro Trimestre da Gravidez/genética , Análise de Sequência de RNA , Primeiro Trimestre da Gravidez/genética , Primeiro Trimestre da Gravidez/metabolismo
7.
Biol Sex Differ ; 15(1): 63, 2024 Aug 16.
Artigo em Inglês | MEDLINE | ID: mdl-39152463

RESUMO

BACKGROUND: Fetal sex and placental development impact pregnancy outcomes and fetal-maternal health, but the critical timepoint of placenta establishment in first trimester is understudied in human pregnancies. METHODS: Pregnant subjects were recruited in late first trimester (weeks 10-14) at time of chorionic villus sampling, a prenatal diagnostic test. Leftover placenta tissue was collected and stored until birth outcomes were known, then DNA and RNA were isolated from singleton, normal karyotype pregnancies resulting in live births. DNA methylation was measured with the Illumina Infinium MethylationEPIC BeadChip array (n = 56). Differential methylation analysis compared 25 females versus 31 males using a generalized linear model on 743,461 autosomal probes. Gene expression sex differences were analyzed with RNA-sequencing (n = 74). An integrated analysis was performed using linear regression to correlate gene expression and DNA methylation in 51 overlapping placentas. RESULTS: Methylation analysis identified 151 differentially methylated probes (DMPs) significant at false discovery rate < 0.05, including 89 (59%) hypermethylated in females. Probe cg17612569 (GABPA, ATP5J) was the most significant CpG site, hypermethylated in males. There were 11 differentially methylated regions affected by fetal sex, with transcription factors ZNF300 and ZNF311 most significantly hypermethylated in males and females, respectively. RNA-sequencing identified 152 genes significantly sexually dimorphic at false discovery rate < 0.05. The 151 DMPs were associated with 18 genes with gene downregulation (P < 0.05) in the direction of hypermethylation, including 2 genes significant at false discovery rate < 0.05 (ZNF300 and CUB and Sushi multiple domains 1, CSMD1). Both genes, as well as Family With Sequence Similarity 228 Member A (FAM228A), showed significant correlation between DNA methylation and sexually dimorphic gene expression, though FAM228A DNA methylation was less sexually dimorphic. Comparison with other sex differences studies found that cg17612569 is male-hypermethylated across gestation in placenta and in human blood up to adulthood. CONCLUSIONS: Overall, sex dimorphic differential methylation with associated differential gene expression in the first trimester placenta is small, but there remain significant genes that may be regulated through methylation leading to differences in the first trimester placenta.


Fetal sex and placenta development affect pregnancy outcomes for both the fetus and mother throughout pregnancy, including risk of miscarriages, preterm birth, preeclampsia, and other outcomes. Epigenetics, the "overlay" of regulatory signals on DNA which affects how DNA is read, is not well understood in early pregnancy when critical placenta developments are happening that affect the rest of pregnancy. Here, we use leftover placenta biopsy samples (n = 56) donated by Cedars-Sinai patients with informed consent to learn about first trimester human placenta DNA methylation differences due to fetal sex. Out of the total 743,461 sites analyzed, we identified 151 sites significantly affected by fetal sex after correcting p-values to reduce false positives (false discovery rate < 0.05). We also performed an analysis to look at multiple sites and identified 11 regions across the genome with significant DNA methylation changes due to fetal sex. Furthermore, because DNA methylation is a regulatory mark on DNA which typically dampens gene expression, we also compared the DNA methylation sex differences to placental RNA-sequencing gene expression analysis using the same tissue from a mostly overlapping patient group (n = 74 total sequenced, n = 51 overlap). We identify 18 genes which show both significant DNA methylation differences and gene expression changes. The most significant gene was transcription factor ZNF300 with higher DNA methylation in males and reduced gene expression in males (and thus higher gene expression in females). This study identifies some sex differences that continue until later pregnancy and others that are unique to first trimester.


Assuntos
Metilação de DNA , Placenta , Primeiro Trimestre da Gravidez , Caracteres Sexuais , Humanos , Feminino , Gravidez , Masculino , Placenta/metabolismo , Adulto
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA