Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 37
Filtrar
1.
Proc Natl Acad Sci U S A ; 120(28): e2217405120, 2023 07 11.
Artículo en Inglés | MEDLINE | ID: mdl-37406095

RESUMEN

Early placenta development involves cytotrophoblast differentiation into extravillous trophoblast (EVT) and syncytiotrophoblast (STB). Defective trophoblast development and function may result in severe pregnancy complications, including fetal growth restriction and pre-eclampsia. The incidence of these complications is increased in pregnancies of fetuses affected by Rubinstein-Taybi syndrome, a developmental disorder predominantly caused by heterozygous mutations in CREB-binding protein (CREBBP) or E1A-binding protein p300 (EP300). Although the acetyltransferases CREBBP and EP300 are paralogs with many overlapping functions, the increased incidence of pregnancy complications is specific for EP300 mutations. We hypothesized that these complications have their origin in early placentation and that EP300 is involved in that process. Therefore, we investigated the role of EP300 and CREBBP in trophoblast differentiation, using human trophoblast stem cells (TSCs) and trophoblast organoids. We found that pharmacological CREBBP/EP300 inhibition blocks differentiation of TSCs into both EVT and STB lineages, and results in an expansion of TSC-like cells under differentiation-inducing conditions. Specific targeting by RNA interference or CRISPR/Cas9-mediated mutagenesis demonstrated that knockdown of EP300 but not CREBBP, inhibits trophoblast differentiation, consistent with the complications seen in Rubinstein-Taybi syndrome pregnancies. By transcriptome sequencing, we identified transforming growth factor alpha (TGFA, encoding TGF-α) as being strongly upregulated upon EP300 knockdown. Moreover, supplementing differentiation medium with TGF-α, which is a ligand for the epidermal growth factor receptor (EGFR), likewise affected trophoblast differentiation and resulted in increased TSC-like cell proliferation. These findings suggest that EP300 facilitates trophoblast differentiation by interfering with at least EGFR signaling, pointing towards a crucial role for EP300 in early human placentation.


Asunto(s)
Preeclampsia , Síndrome de Rubinstein-Taybi , Embarazo , Femenino , Humanos , Trofoblastos/metabolismo , Factor de Crecimiento Transformador alfa , Síndrome de Rubinstein-Taybi/genética , Síndrome de Rubinstein-Taybi/metabolismo , Diferenciación Celular , Proteína p300 Asociada a E1A/genética , Proteína de Unión a CREB/genética , Receptores ErbB
2.
Prenat Diagn ; 42(13): 1612-1621, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-36336875

RESUMEN

OBJECTIVE: Noninvasive Prenatal Diagnosis has recently been introduced for a limited number of monogenetic disorders. However, the majority of DNA diagnostics still require fetal material obtained using an invasive test. Recently, a novel technique, TRIC (Trophoblast Retrieval and Isolation from the Cervix), has been described, which collects fetal trophoblast cells by endocervical sampling. Since this technique has not been successfully replicated by other groups, we aimed to achieve this in the current study. METHOD: Pregnant women referred for transvaginal chorionic villous sampling (CVS) were asked for an endocervical sample prior to CVS. The TRIC samples were processed to isolate trophoblast DNA. TRIC DNA was used in ForenSeq to determine the amount of maternal DNA contamination, and for Sanger sequencing in case of a monogenic disorder. RESULTS: 23%-44% of samples had a sufficiently high fetal DNA fraction to allow genetic testing, as calculated by Sanger sequencing and ForenSeq, respectively. CONCLUSION: We have been able to successfully replicate the TRIC protocol, although with a much lower success rate as described by the original study performing TRIC. As we obtained the samples in the actual clinical setting envisioned, the method in its current setup is not advisable for use in prenatal diagnostics.


Asunto(s)
Cuello del Útero , Trofoblastos , Embarazo , Femenino , Humanos , Diagnóstico Prenatal/métodos , Pruebas Genéticas , Muestra de la Vellosidad Coriónica
3.
Placenta ; 125: 2-3, 2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-34656364
4.
Front Cell Dev Biol ; 9: 671806, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34095140

RESUMEN

Mutations in the LINC-HELLP non-coding RNA (HELLPAR) have been associated with familial forms of the pregnancy-specific HELLP syndrome. These mutations negatively affect extravillous trophoblast (EVT) differentiation from a proliferative to an invasive state and disturb the binding of RNA splicing complex proteins PCBP1, PCBP2, and YBX1 to LINC-HELLP. In this study, by using both in vitro and ex vivo experiments, we investigate if these proteins are involved in the regulation of EVT invasion during placentation. Additionally, we study if this regulation is due to alternative mRNA splicing. HTR-8/SVneo extravillous trophoblasts and human first trimester placental explants were used to investigate the effect of siRNA-mediated downregulation of PCBP1, PCBP2, and YBX1 genes on the differentiation of EVTs. Transwell invasion assays and proliferation assays indicated that upon knockdown of PCBP2 and, to a lesser extent, YBX1 and PCBP1, EVTs fail to differentiate toward an invasive phenotype. The same pattern was observed in placental explants where PCBP2 knockdown led to approximately 80% reduction in the number of explants showing any EVT outgrowth. Of the ones that still did show EVT outgrowth, the percentage of proliferating EVTs was significantly higher compared to explants transfected with non-targeting control siRNAs. To further investigate this effect of PCBP2 silencing on EVTs, we performed whole transcriptome sequencing (RNA-seq) on HTR-8/SVneo cells after PCBP2 knockdown. PCBP2 knockdown was found to have minimal effect on mRNA expression levels. In contrast, PCBP2 silencing led to a switch in splicing for a large number of genes with predominant functions in cellular assembly and organization, cellular function and maintenance, and cellular growth and proliferation and the cell cycle. EVTs, upon differentiation, alter their function to be able to invade the decidua of the mother by changing their cellular assembly and their proliferative activity by exiting the cell cycle. PCBP2 appears to be a paramount regulator of these differentiation mechanisms, where its disturbed binding to LINC-HELLP could contribute to dysfunctional placental development as seen in the HELLP syndrome.

5.
Hypertension ; 77(2): 475-490, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33356399

RESUMEN

Revaluation of the association of the STOX1 (STORKHEAD_BOX1 PROTEIN 1) transcription factor mutation (Y153H, C allele) with the early utero-vascular origins of placental pathology is warranted. To investigate if placental STOX1 Y153H genotype affects utero-vascular remodeling-compromised in both preterm birth and preeclampsia-we utilized extravillous trophoblast (EVT) explant and placental decidual coculture models, transfection of STOX1 wild-type and mutant plasmids into EVT-like trophoblast cell lines, and a cohort of 75 placentas from obstetric pathologies. Primary EVT and HTR8/SVneo cells carrying STOX1 Y153H secreted lower levels of IL (interleukin) 6, and IL-8, and higher CXCL16 (chemokine [C-X-C motif] ligand 16) and TRAIL (tumor necrosis factor-related apoptosis-inducing ligand) than wild-type EVT and Swan71 cells. Media from wild-type EVT or Swan71 cells transfected with wild-type STOX1 stimulated: endothelial chemokine expression, angiogenesis, and decidual natural killer cell and monocyte migration. In contrast, Y153H EVT conditioned medium, Swan71 transfected with the Y153H plasmid, or HTR8/SVneo media had no effect. Genotyping of placental decidual cocultures demonstrated association of the placental STOX1 CC allele with failed vascular remodeling. Decidual GG NODAL R165H increased in failed cocultures carrying the placental CC alleles of STOX1. Multivariate analysis of the placental cohort showed that the STOX1 C allele correlated with premature birth, with or without severe early-onset preeclampsia, and small for gestational age babies. In conclusion, placental STOX1 Y153H is a precipitating factor in preterm birth and placental preeclampsia due to defects in early utero-placental development.


Asunto(s)
Proteínas Portadoras/metabolismo , Placenta/metabolismo , Placentación/genética , Preeclampsia/metabolismo , Nacimiento Prematuro/metabolismo , Trofoblastos/metabolismo , Adolescente , Adulto , Alelos , Proteínas Portadoras/genética , Línea Celular , Femenino , Humanos , Recién Nacido , Interleucina-6/metabolismo , Interleucina-8/metabolismo , Preeclampsia/genética , Embarazo , Primer Trimestre del Embarazo/metabolismo , Nacimiento Prematuro/genética , Adulto Joven
7.
Sci Rep ; 9(1): 19077, 2019 12 13.
Artículo en Inglés | MEDLINE | ID: mdl-31836787

RESUMEN

Preeclampsia is a frequent gestational hypertensive disorder with equivocal pathophysiology. Knockout of peptide hormone ELABELA (ELA) has been shown to cause preeclampsia-like symptoms in mice. However, the role of ELA in human placentation and whether ELA is involved in the development of preeclampsia in humans is not yet known. In this study, we show that exogenous administration of ELA peptide is able to increase invasiveness of extravillous trophoblasts in vitro, is able to change outgrowth morphology and reduce trophoblast proliferation ex vivo, and that these effects are, at least in part, independent of signaling through the Apelin Receptor (APLNR). Moreover, we show that circulating levels of ELA are highly variable between women, correlate with BMI, but are significantly reduced in first trimester plasma of women with a healthy BMI later developing preeclampsia. We conclude that the large variability and BMI dependence of ELA levels in circulation make this peptide an unlikely candidate to function as a first trimester preeclampsia screening biomarker, while in the future administering ELA or a derivative might be considered as a potential preeclampsia treatment option as ELA is able to drive extravillous trophoblast differentiation.


Asunto(s)
Diferenciación Celular , Hormonas Peptídicas/sangre , Placenta/metabolismo , Trofoblastos/citología , Adulto , Apelina/sangre , Índice de Masa Corporal , Línea Celular , Proliferación Celular , Estudios de Cohortes , Femenino , Humanos , Preeclampsia/sangre , Embarazo , Primer Trimestre del Embarazo/sangre , Gemelos
8.
Pregnancy Hypertens ; 17: 148-157, 2019 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-31487633

RESUMEN

The apelinergic-axis (Apelin, Elabela and their receptor APJ) is involved in many physiological and pathological processes. Both Elabela/APJ and Apelin/APJ are implicated in the pathophysiology of preeclampsia in rodents. However, the findings regarding the apelinergic axis in human preeclamptic placental development have been rather conflicting. In this systematic review we present an overview of the current evidence regarding the pathophysiological role of Apelin, Elabela and their receptor in human preeclamptic pregnancies. The databases used for this systematic review were Pubmed, Scopus, Google Scholar and ClinicalTrials.gov. The reference lists of the selected studies were also screened for any additional studies. The last search was performed on the 25th of March 2019. Thirteen studies were included and subjected to quality assessment so that only high quality datasets were finally selected and included in this systematic evaluation. In total, 410 women that developed preeclampsia or IUGR and 409 healthy control pregnancies were included. The findings of this review suggest that circulating Apelin levels are increased in early onset/severe preeclamptic patients while Apelin levels in severe preeclamptic placenta tissues appear to show the opposite. Circulating Elabela levels in early-onset preeclamptic women do not differ from controls, while its levels in late-onset preeclampsia remain inconclusive. The studies on Elabela and APJ expression in placental tissues require larger sample sizes with defined preeclampsia subtypes to draw any definite conclusions. Large cohort studies with affected and control groups matched for Body Mass Index and gestational age at sampling are essential in order to substantiate other current findings.


Asunto(s)
Receptores de Apelina/metabolismo , Apelina/metabolismo , Hormonas Peptídicas/metabolismo , Placenta/metabolismo , Preeclampsia/metabolismo , Femenino , Humanos , Preeclampsia/sangre , Embarazo
9.
PLoS One ; 13(8): e0202190, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30092105

RESUMEN

The number of molecules identified to be involved in communication between placenta and decidua is fast expanding. Previously, we showed that NODAL expressed in maternal endometrial stromal cells is able to affect NODAL and STOX1 expression in placental extravillous trophoblasts. The effect of maternal NODAL on placental NODAL expression is achieved via Activin A, while preliminary data suggests that maternal NODAL affects STOX1 expression in trophoblasts potentially via IGF1. In the current study, T-HESC endometrial stromal cells were treated with siRNAs against NODAL after which IGF1 mRNA expression was determined by quantitative RT-PCR, while IGF1 secretion was measured by ELISA. Recombinant IGF1 and inhibitors of the MAPK and PI3K/AKT pathways were added to SGHPL-5 extravillous trophoblasts after which the effects on STOX1 mRNA and STOX1 protein expression were determined. The effect of IGF1 and the MAPK and PI3K/AKT inhibitors on the invasive capacity of SGHPL-5 cells was investigated by performing invasion assays. We found that T-HESC cells treated with NODAL siRNAs showed significant upregulation of IGF1 mRNA expression and IGF1 protein secretion. Addition of IGF1 to SGHPL-5 cell media significantly upregulated STOX1 mRNA and protein expression. Using inhibitors of the PI3K/AKT and MAPK pathway showed that the effect of IGF1 on STOX1 expression is accomplished via MAPK signaling. Secondly, PI3K inhibition independently leads to reduced STOX1 expression which can be rescued by adding IGF1. IGF1 was unable to influence the invasive capacity of SGHPL-5 cells, while inhibiting the PI3K/AKT pathway did reduce the invasion of these cells. To conclude, here we show that downregulated NODAL expression in endometrial stromal cells, previously associated with pre-eclampsia like symptoms in mice, increases IGF1 secretion. Increased levels of IGF1 lead to increased expression levels of STOX1 in extravillous trophoblasts via the MAPK pathway, hereby identifying a novel signaling cascade involved in maternal-fetal communication.


Asunto(s)
Proteínas Portadoras/metabolismo , Factor I del Crecimiento Similar a la Insulina/metabolismo , Proteína Nodal/metabolismo , Trofoblastos/metabolismo , Línea Celular , Endometrio/metabolismo , Femenino , Regulación del Desarrollo de la Expresión Génica , Humanos , Proteínas Quinasas Activadas por Mitógenos/antagonistas & inhibidores , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Proteína Nodal/antagonistas & inhibidores , Proteína Nodal/genética , Fosfatidilinositol 3-Quinasas/metabolismo , Inhibidores de las Quinasa Fosfoinosítidos-3 , Proteínas Proto-Oncogénicas c-akt/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-akt/metabolismo , ARN Mensajero/metabolismo , Proteínas Recombinantes/metabolismo , Células del Estroma/metabolismo
10.
J Neuroinflammation ; 14(1): 235, 2017 Dec 04.
Artículo en Inglés | MEDLINE | ID: mdl-29202778

RESUMEN

BACKGROUND: Multiple sclerosis (MS) is a demyelinating and degenerative disease of the central nervous system. Normally, demyelination is followed by remyelination, which requires repopulation of a demyelinated area by oligodendrocyte precursor cells. Although large numbers of precursor cells are present in MS lesions, remyelination often fails, in part by the inability of precursor cells to differentiate into mature myelin-forming cells. In mouse and rat, miR-219 is required for this differentiation. Previously, we identified decreased miR-219 expression in tissue of MS patients compared to controls. Cell-free miRNAs have been detected in many different body fluids including cerebrospinal fluid (CSF) and may reflect disease processes going on in the central nervous system. This prompted us to investigate the biomarker performance of CSF miR-219 for MS diagnosis. METHODS: Quantitative PCR was performed measuring miR-219 levels in CSF of MS patients and controls in three independent cohorts. RESULTS: All three cohorts of MS patients and controls revealed that absence of miR-219 detection in CSF is consistently associated with MS. CONCLUSIONS: We have been able to identify and validate absence of miR-219 detection in CSF of MS patients compared to controls, suggesting that it may emerge as a candidate biomarker for MS diagnosis.


Asunto(s)
Biomarcadores/líquido cefalorraquídeo , MicroARNs/líquido cefalorraquídeo , Esclerosis Múltiple/líquido cefalorraquídeo , Adulto , Femenino , Humanos , Masculino , Persona de Mediana Edad
11.
Science ; 357(6352): 707-713, 2017 08 18.
Artículo en Inglés | MEDLINE | ID: mdl-28663440

RESUMEN

Preeclampsia (PE) is a gestational hypertensive syndrome affecting between 5 and 8% of all pregnancies. Although PE is the leading cause of fetal and maternal morbidity and mortality, its molecular etiology is still unclear. Here, we show that ELABELA (ELA), an endogenous ligand of the apelin receptor (APLNR, or APJ), is a circulating hormone secreted by the placenta. Elabela but not Apelin knockout pregnant mice exhibit PE-like symptoms, including proteinuria and elevated blood pressure due to defective placental angiogenesis. In mice, infusion of exogenous ELA normalizes hypertension, proteinuria, and birth weight. ELA, which is abundant in human placentas, increases the invasiveness of trophoblast-like cells, suggesting that it enhances placental development to prevent PE. The ELA-APLNR signaling axis may offer a new paradigm for the treatment of common pregnancy-related complications, including PE.


Asunto(s)
Anomalías Cardiovasculares/genética , Proteínas Portadoras/genética , Hormonas Placentarias/genética , Placentación/genética , Preeclampsia/genética , Animales , Apelina/genética , Apelina/metabolismo , Peso al Nacer , Proteínas Portadoras/administración & dosificación , Proteínas Portadoras/metabolismo , Proteínas Portadoras/farmacología , Femenino , Ratones , Ratones Noqueados , Neovascularización Fisiológica/genética , Hormonas Peptídicas , Placenta/irrigación sanguínea , Placenta/metabolismo , Embarazo , Proteinuria , Transducción de Señal
12.
Sci Transl Med ; 8(363): 363re4, 2016 Nov 02.
Artículo en Inglés | MEDLINE | ID: mdl-27807286

RESUMEN

Single-gene mutations account for more than 6000 diseases, 10% of all pediatric hospital admissions, and 20% of infant deaths. Down syndrome and other aneuploidies occur in more than 0.2% of births worldwide and are on the rise because of advanced reproductive age. Birth defects of genetic origin can be diagnosed in utero after invasive extraction of fetal tissues. Noninvasive testing with circulating cell-free fetal DNA is limited by a low fetal DNA fraction. Both modalities are unavailable until the end of the first trimester. We have isolated intact trophoblast cells from Papanicolaou smears collected noninvasively at 5 to 19 weeks of gestation for next-generation sequencing of fetal DNA. Consecutive matched maternal, placental, and fetal samples (n = 20) were profiled by multiplex targeted DNA sequencing of 59 short tandem repeat and 94 single-nucleotide variant sites across all 24 chromosomes. The data revealed fetal DNA fractions of 85 to 99.9%, with 100% correct fetal haplotyping. This noninvasive platform has the potential to provide comprehensive fetal genomic profiling as early as 5 weeks of gestation.


Asunto(s)
Feto/patología , Mutación , Diagnóstico Prenatal/métodos , Trofoblastos/citología , Ácidos Nucleicos Libres de Células/análisis , Análisis Mutacional de ADN , Femenino , Genotipo , Edad Gestacional , Haplotipos , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Repeticiones de Microsatélite , Placenta/metabolismo , Polimorfismo de Nucleótido Simple , Embarazo , Primer Trimestre del Embarazo , Atención Prenatal
13.
Sci Rep ; 6: 32129, 2016 08 24.
Artículo en Inglés | MEDLINE | ID: mdl-27555360

RESUMEN

The familial forms of early onset pre-eclampsia and related syndromes (HELLP) present with hypertension and proteinuria in the mother and growth restriction of the fetus. Genetically, these clinically similar entities are caused by different founder-dependent, placentally-expressed paralogous genes. All susceptibility genes (STOX1, lincHELLP, INO80B) identified so far are master control genes that regulate an essential trophoblast differentiation pathway, but act at different entry points. Many genes remain to be identified. Here we demonstrate that a long non-coding RNA (lncRNA) within intron 3 of the STOX2 gene on 4q35.1 acts as a permissive cis-acting regulator of alternative splicing of STOX2. When this lncRNA is mutated or absent, an alternative exon (3B) of STOX2 is included. This introduces a stop codon resulting in the deletion of a highly conserved domain of 64 amino acids in the C-terminal of the STOX2 protein. A mutation present within a regulatory region within intron 1 of STOX2 has the same effect after blocking with CRISPR technology: transcripts with exon 3B are upregulated. This proces appears related to transcriptional control by a chromatin-splicing adaptor complex as described for FGFR2. For STOX2, CHD5, coding for a chromodomain helicase DNA binding protein, qualifies as the chromatin modifier in this process.


Asunto(s)
Proteínas Portadoras/genética , Cromosomas Humanos Par 4 , Preeclampsia/genética , ARN no Traducido/genética , Empalme Alternativo , Sistemas CRISPR-Cas , Femenino , Finlandia , Predisposición Genética a la Enfermedad , Humanos , Intrones , Masculino , Mutación , Linaje , Placenta/citología , Placenta/fisiología , Polimorfismo de Nucleótido Simple , Embarazo , Trofoblastos/patología
14.
Cell Death Differ ; 23(12): 2042-2053, 2016 12.
Artículo en Inglés | MEDLINE | ID: mdl-27564589

RESUMEN

Cerebellar granule neuronal progenitors (GNPs) are the precursors of cerebellar granule cells (CGCs) and are believed to be the cell of origin for medulloblastoma (MB), yet the molecular mechanisms governing GNP neurogenesis are poorly elucidated. Here, we demonstrate that storkhead box 1 (Stox1), a forkhead transcriptional factor, has a pivotal role in cerebellar granule neurogenesis and MB suppression. Expression of Stox1 is upregulated along with GNP differentiation and repressed by activation of sonic hedgehog (SHH) signaling. Stox1 exerts its neurogenic and oncosuppressing effect via direct transcriptional repression of Math1, a basic helix-loop-helix transcription activator essential for CGC genesis. This study illustrates a SHH-Stox1-Math1 regulatory axis in normal cerebellar development and MB formation.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Proteínas Portadoras/metabolismo , Neoplasias Cerebelosas/genética , Cerebelo/metabolismo , Gránulos Citoplasmáticos/metabolismo , Meduloblastoma/genética , Neurogénesis , Transcripción Genética , Animales , Secuencia de Bases , Diferenciación Celular , Proliferación Celular , Neoplasias Cerebelosas/patología , Cerebelo/crecimiento & desarrollo , Técnicas de Silenciamiento del Gen , Proteínas Hedgehog , Meduloblastoma/patología , Ratones , Células-Madre Neurales/citología , Células-Madre Neurales/metabolismo , Receptores Patched/deficiencia , Receptores Patched/metabolismo , Transducción de Señal
15.
PLoS One ; 11(2): e0148313, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26870946

RESUMEN

BACKGROUND: The physiological demands of pregnancy on the maternal cardiovascular system can catapult women into a metabolic syndrome that predisposes to atherosclerosis in later life. We sought to identify the nature of the epigenomic changes associated with the increased cardiovascular disease (CVD) risk in adult women following pre-eclampsia. FINDINGS: We assessed the genome wide epigenetic profile by methyl-C sequencing of monozygotic parous twin sister pairs discordant for a severe variant of pre-eclampsia. In the adult twin sisters at risk for CVD as a consequence of a complicated pregnancy, a set of 12 differentially methylated regions with at least 50% difference in methylation percentage and the same directional change was found to be shared between the affected twin sisters and significantly different compared to their unaffected monozygous sisters. CONCLUSION: The current epigenetic marker set will permit targeted analysis of differentially methylated regions potentially related to CVD risk in large cohorts of adult women following complicated pregnancies.


Asunto(s)
Enfermedades Cardiovasculares/genética , Islas de CpG , Epigénesis Genética , Genoma Humano , Preeclampsia/genética , Adulto , Enfermedades Cardiovasculares/etiología , Enfermedades Cardiovasculares/fisiopatología , Sistema Cardiovascular/metabolismo , Sistema Cardiovascular/fisiopatología , Metilación de ADN , Femenino , Marcadores Genéticos , Estudio de Asociación del Genoma Completo , Humanos , Estudios Longitudinales , Preeclampsia/fisiopatología , Embarazo , Factores de Riesgo , Índice de Severidad de la Enfermedad , Hermanos , Factores de Tiempo , Gemelos Monocigóticos/genética
16.
Hum Mol Genet ; 24(19): 5475-85, 2015 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-26173455

RESUMEN

LINC-HELLP, showing chromosomal linkage with the pregnancy-specific HELLP syndrome in Dutch families, reduces differentiation from a proliferative to an invasive phenotype of first-trimester extravillous trophoblasts. Here we show that mutations in LINC-HELLP identified in HELLP families negatively affect this trophoblast differentiation either by inducing proliferation rate or by causing cell cycle exit as shown by a decrease in both proliferation and invasion. As LincRNAs predominantly function through interactions with proteins, we identified the directly interacting proteins using chromatin isolation by RNA purification followed by protein mass spectrometry. We found 22 proteins predominantly clustering in two functional networks, i.e. RNA splicing and the ribosome. YBX1, PCBP1, PCBP2, RPS6 and RPL7 were validated, and binding to these proteins was influenced by the HELLP mutations carried. Finally, we show that the LINC-HELLP transcript levels are significantly upregulated in plasma of women in their first trimester of pregnancy compared with non-pregnant women, whereas this upregulation seems absent in a pilot set of patients later developing pregnancy complications, indicative of its functional significance in vivo.


Asunto(s)
Síndrome HELLP/genética , Mutación , Primer Trimestre del Embarazo/genética , ARN Largo no Codificante/genética , Trofoblastos/citología , Ciclo Celular , Diferenciación Celular , Proliferación Celular , Femenino , Regulación de la Expresión Génica , Síndrome HELLP/sangre , Humanos , Embarazo , Primer Trimestre del Embarazo/sangre , Proteínas/metabolismo , Empalme del ARN , ARN Largo no Codificante/sangre , Ribosomas/metabolismo
17.
Hum Mol Genet ; 24(1): 118-27, 2015 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-25143393

RESUMEN

In humans, the elucidation of the genetics underlying multifactorial diseases such as pre-eclampsia remains complex. Given the current day availability of genome-wide linkage- and expression data pools, we applied pathway-guided genome-wide meta-analysis guided by the premise that the functional network underlying these multifactorial syndromes is under selective genetic pressure. This approach drastically reduced the genomic region of interest, i.e. 2p13 linked with pre-eclampsia in Icelandic families, from 8 679 641 bp (region with linkage) to 45 264 bp (coding exons of prioritized genes) (0.83%). Mutation screening of the candidate genes (n = 13) rapidly reduced the minimal critical region and showed the INO80B gene, encoding a novel winged helix domain (pfam14465) and part of the chromatin-remodeling complex, to be linked to pre-eclampsia. The functional defect in placental cells involved a susceptibility allele-dependent loss-of-gene silencing due to increased INO80B RNA stability as a consequence of differential binding of miR-1324 to the susceptibility allele of rs34174194. This risk allele is located at position 1 in an absolutely conserved 7-mer (UUGUCUG) in the 3-UTR of INO80B immediately downstream of a variant Pumillio Recognition Element (UGUANAAG). These data support that pre-eclampsia genes affect a conserved fundamental mechanism that evolved as a consequence of hemochorial placentation. Functionally, this involves founder-dependent, placentally expressed paralogous genes that regulate an essential trophoblast differentiation pathway but act at different entry points.


Asunto(s)
Proteínas de Ciclo Celular/genética , Estudios de Asociación Genética/métodos , MicroARNs/genética , Proteínas Nucleares/genética , Preeclampsia/genética , Análisis Mutacional de ADN/métodos , Femenino , Predisposición Genética a la Enfermedad , Humanos , Islandia , Péptidos y Proteínas de Señalización Intracelular/genética , Placenta/metabolismo , Polimorfismo de Nucleótido Simple , Embarazo
18.
Front Genet ; 5: 38, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24596577
19.
Front Genet ; 4: 180, 2013 Sep 10.
Artículo en Inglés | MEDLINE | ID: mdl-24058367

RESUMEN

This review describes the current knowledge regarding genetics and epigenetics of pregnancy-associated diseases with placental origin. We discuss the effect on genetic linkage analyses when the fetal genotype determines the maternal phenotype. Secondly, the genes identified by genome-wide linkage studies to be associated with pre-eclampsia (ACVR2A, STOX1) and the HELLP-syndrome (LINC-HELLP) are discussed regarding their potential functions in the etiology of disease. Furthermore, susceptibility genes identified by candidate gene approaches (e.g., CORIN) are described. Next, we focus on the additional challenges that come when epigenetics also play a role in disease inheritance. We discuss the maternal transmission of the chromosome 10q22 pre-eclampsia linkage region containing the STOX1 gene and provide further evidence for the role of epigenetics in pre-eclampsia based on the cdkn1c mouse model of pre-eclampsia. Finally, we provide recommendations to unravel the genetics of pregnancy-associated diseases, specifically regarding clear definitions of patient groups and sufficient patient numbers, and the potential usefulness of (epi)genetic data in early non-invasive biomarker development.

20.
Front Genet ; 4: 170, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23986775

RESUMEN

Nodal, a secreted signaling protein from the transforming growth factor beta (TGF-ß)-super family plays a vital role during early embryonic development. Recently, it was found that maternal decidua-specific Nodal knockout mice show intrauterine growth restriction (IUGR) and preterm birth. The chromosomal location of NODAL is in the same linkage area as the placental (fetal) pre-eclampsia (PE) susceptibility gene STOX1, which is associated with the familial form of early-onset, IUGR-complicated PE. As the STOX1 linkage was originally identified in women being born from a pre-eclamptic pregnancy as well as suffering from PE themselves, the linkage could in part be caused by NODAL, which is why the potential maternal-fetal interaction between STOX1 and NODAL was investigated. In the PE families with the STOX1 susceptibility allele carried by the children born from pre-eclamptic pregnancies, it was found that the pre-eclamptic mothers themselves all carried the NODAL H165R SNP, which causes a 50% reduced activity. Surprisingly, in decidua-specific Nodal knockout mice the fetal placenta showed up-regulation of STOX1 and NODAL expression. Conditioned media of human first trimester decidua and a human endometrial stromal cell line (T-HESC) treated with siRNAs against NODAL or carrying the H165R SNP were also able to induce NODAL and STOX1 expression when added to SGHPL-5 first trimester extravillous trophoblast cells. Finally, a human TGF-ß/BMP signaling pathway PCR-array on decidua and the T-HESC cell line with Nodal knockdown revealed upregulation of Activin-A, which was confirmed in conditioned media by ELISA. We show that maternal decidua Nodal knockdown gives upregulation of NODAL and STOX1 mRNA expression in fetal extravillous trophoblast cells, potentially via upregulation of Activin-A in the maternal decidua. As both Activin-A and Nodal have been implicated in PE, being increased in serum of pre-eclamptic women and upregulated in pre-eclamptic placentas respectively, this interaction at the maternal-fetal interface might play a substantial role in the development of PE.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...