Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 36
Filtrar
1.
Cell Mol Biol Lett ; 29(1): 74, 2024 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-38750428

RESUMEN

By analyzing a mouse Interspecific Recombinant Congenic Strain (IRCS), we previously identified a quantitative trait locus (QTL), called Mafq1 on mouse chromosome 1, that is associated with male hypofertility and ultrastructural sperm abnormalities. Within this locus, we identified a new candidate gene that could be implicated in a reproductive phenotype: Tex44 (Testis-expressed protein 44). We thus performed a CRISPR/Cas9-mediated complete deletion of this gene in mice in order to study its function. Tex44-KO males were severely hypofertile in vivo and in vitro due to a drastic reduction of sperm motility which itself resulted from important morphological sperm abnormalities. Namely, Tex44-KO sperm showed a disorganized junction between the midpiece and the principal piece of the flagellum, leading to a 180° flagellar bending in this region. In addition, the loss of some axonemal microtubule doublets and outer dense fibers in the flagellum's principal piece has been observed. Our results suggest that, in mice, TEX44 is implicated in the correct set-up of the sperm flagellum during spermiogenesis and its absence leads to flagellar abnormalities and consequently to severe male hypofertility.


Asunto(s)
Infertilidad Masculina , Ratones Noqueados , Motilidad Espermática , Cola del Espermatozoide , Animales , Masculino , Infertilidad Masculina/genética , Infertilidad Masculina/patología , Motilidad Espermática/genética , Cola del Espermatozoide/patología , Cola del Espermatozoide/metabolismo , Ratones , Espermatozoides/metabolismo , Espermatogénesis/genética , Flagelos/genética , Flagelos/metabolismo , Ratones Endogámicos C57BL , Sistemas CRISPR-Cas/genética
2.
Int J Mol Sci ; 23(22)2022 Nov 09.
Artículo en Inglés | MEDLINE | ID: mdl-36430291

RESUMEN

Fertilization is a complex process that requires successive stages and culminates in the adhesion/fusion of gamete membranes. If the question of the involvement of oocyte integrins has been swept away by deletion experiments, that of the involvement of sperm integrins remains to be further characterized. In the present study, we addressed the question of the feasibility of sperm-oocyte adhesion/fusion and early implantation in the absence of sperm ß1 integrin. Males and females with ß1 integrin-depleted sperm and oocytes were mated, and fertilization outcome was monitored by a gestational ultrasound analysis. Results suggest that although the sperm ß1 integrin participates in gamete adhesion/fusion, it is dispensable for fertilization in mice. However, sperm- and/or oocyte-originated integrin ß1 is essential for post-implantation development. Redundancy phenomena could be at the origin of a compensatory expression or alternative dimerization pattern.


Asunto(s)
Integrina beta1 , Interacciones Espermatozoide-Óvulo , Femenino , Ratones , Masculino , Animales , Integrina beta1/genética , Integrina beta1/metabolismo , Semen/metabolismo , Oocitos/metabolismo , Espermatozoides/metabolismo , Fertilización , Integrinas/metabolismo
3.
Int J Mol Sci ; 22(4)2021 Feb 16.
Artículo en Inglés | MEDLINE | ID: mdl-33669425

RESUMEN

Thanks to the analysis of an Interspecific Recombinant Congenic Strain (IRCS), we previously defined the Mafq1 quantitative trait locus as an interval on mouse Chromosome 1 associated with male hypofertility and ultrastructural abnormalities. We identified the Spermatogenesis associated protein 3 gene (Spata3 or Tsarg1) as a pertinent candidate within the Mafq1 locus and performed the CRISPR-Cas9 mediated complete deletion of the gene to investigate its function. Male mice deleted for Spata3 were normally fertile in vivo but exhibited a drastic reduction of efficiency in in vitro fertilization assays. Mobility parameters were normal but ultrastructural analyses revealed acrosome defects and an overabundance of lipids droplets in cytoplasmic remnants. The deletion of the Spata3 gene reproduces therefore partially the phenotype of the hypofertile IRCS strain.


Asunto(s)
Acrosoma/patología , Fertilización In Vitro/métodos , Eliminación de Gen , Infertilidad Masculina/genética , Proteínas/genética , Acrosoma/metabolismo , Acrosoma/ultraestructura , Animales , Sistemas CRISPR-Cas , Modelos Animales de Enfermedad , Femenino , Infertilidad Masculina/metabolismo , Gotas Lipídicas/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Fenotipo , Embarazo , Proteínas/metabolismo , Motilidad Espermática/genética , Espermatogénesis/genética , Testículo/metabolismo
4.
Int J Mol Sci ; 21(22)2020 Nov 12.
Artículo en Inglés | MEDLINE | ID: mdl-33198087

RESUMEN

Male fertility disorders often have their origin in disturbed spermatogenesis, which can be induced by genetic factors. In this study, we used interspecific recombinant congenic mouse strains (IRCS) to identify genes responsible for male infertility. Using ultrasonography, in vivo and in vitro fertilization (IVF) and electron microscopy, the phenotyping of several IRCS carrying mouse chromosome 1 segments of Mus spretus origin revealed a decrease in the ability of sperm to fertilize. This teratozoospermia included the abnormal anchoring of the acrosome to the nucleus and a persistence of residual bodies at the level of epididymal sperm midpiece. We identified a quantitative trait locus (QTL) responsible for these phenotypes and we have proposed a short list of candidate genes specifically expressed in spermatids. The future functional validation of candidate genes should allow the identification of new genes and mechanisms involved in male infertility.


Asunto(s)
Cromosomas Humanos Par 1/genética , Infertilidad Masculina/genética , Sitios de Carácter Cuantitativo/genética , Acrosoma/fisiología , Animales , Núcleo Celular/genética , Núcleo Celular/fisiología , Epidídimo/fisiología , Femenino , Humanos , Masculino , Ratones , Fenotipo , Espermátides/fisiología , Espermatogénesis/genética , Espermatozoides/fisiología , Teratozoospermia/genética
5.
Int J Mol Sci ; 21(22)2020 Nov 11.
Artículo en Inglés | MEDLINE | ID: mdl-33187358

RESUMEN

We have previously shown, using antibodies, that the sperm alpha6beta1 integrin is involved in mouse gamete fusion in vitro. Here we report the conditional knockdown of the sperm Itgb1 gene. It induced a drastic failure of sperm fusogenic ability with sperm accumulation in the perivitelline space of in vitro inseminated oocytes deleted or not for the Itgb1 gene. These data demonstrate that sperm, but not oocyte, beta1 integrin subunit is involved in gamete adhesion/fusion. Curiously, knockdown males were fertile in vivo probably because of the incomplete Cre-mediated deletion of the sperm Itgb1 floxed gene. Indeed, this was shown by Western blot analysis and confirmed by both the viability and litter size of pups obtained by mating partially sperm Itgb1 deleted males with females producing completely deleted Itgb1 oocytes. Because of the total peri-implantation lethality of Itgb1 deletion in mice, we assume that sperm that escaped the Itgb1 excision seemed to be preferentially used to fertilize in vivo. Here, we showed for the first time that the deletion, even partial, of the sperm Itgb1 gene makes the sperm unable to normally fertilize oocytes. However, to elucidate the question of the essentiality of its role during fertilization, further investigations using a mouse expressing a recombinase more effective in male germ cells are necessary.


Asunto(s)
Adhesión Celular/genética , Células Germinativas/fisiología , Integrina beta1/genética , Subunidades de Proteína/genética , Animales , Adhesión Celular/fisiología , Fusión Celular/métodos , Femenino , Fertilización/genética , Fertilización/fisiología , Masculino , Ratones , Ratones Noqueados , Oocitos/fisiología , Interacciones Espermatozoide-Óvulo/genética , Interacciones Espermatozoide-Óvulo/fisiología , Espermatozoides/fisiología
6.
Sci Rep ; 10(1): 5335, 2020 03 24.
Artículo en Inglés | MEDLINE | ID: mdl-32210282

RESUMEN

Three genes are known to be essential for gamete adhesion/fusion (Cd9, Izumo1 and Juno). Here, we confirmed that Spaca6 null males are infertile and showed that their sperm accumulate in the perivitelline space but are unable to fuse with oocyte. Like IZUMO1, SPACA6 which is expressed by human sperm, is remained on the equatorial segment after acrosomal reaction and is involved in human fertilization since an anti-SPACA6 antibody inhibited it. Despite the similarity of the phenotypes caused by Spaca6 and Izumo1 knockouts, these are not redundant and the essential relocation of IZUMO1 is not affected by the lack of SPACA6. We propose a model in which IZUMO1 and SPACA6 would be part of a molecular complex necessary for gamete fusion and that their concomitant presence would be required for the recruitment of another essential molecular actor, such as a fusogen, for the fusion to take place.


Asunto(s)
Proteínas de Plasma Seminal/metabolismo , Interacciones Espermatozoide-Óvulo/fisiología , Espermatozoides/fisiología , Reacción Acrosómica , Animales , Células COS , Chlorocebus aethiops , Femenino , Fertilización In Vitro , Humanos , Inmunoglobulinas/genética , Inmunoglobulinas/metabolismo , Infertilidad Masculina/genética , Infertilidad Masculina/terapia , Masculino , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Ratones Endogámicos C57BL , Ratones Endogámicos , Ratones Noqueados , Ratones Transgénicos , Proteínas de Plasma Seminal/genética , Cabeza del Espermatozoide/fisiología , Inyecciones de Esperma Intracitoplasmáticas
7.
Mol Med ; 25(1): 37, 2019 08 08.
Artículo en Inglés | MEDLINE | ID: mdl-31395028

RESUMEN

BACKGROUND: Human reproductive disorders consist of frequently occurring dysfunctions including a broad range of phenotypes affecting fertility and women's health during pregnancy. Several female-related diseases have been associated with hypofertility/infertility phenotypes, such as recurrent pregnancy loss (RPL). Other occurring diseases may be life-threatening for the mother and foetus, such as preeclampsia (PE) and intra-uterine growth restriction (IUGR). FOXD1 was defined as a major molecule involved in embryo implantation in mice and humans by regulating endometrial/placental genes. FOXD1 mutations in human species have been functionally linked to RPL's origin. METHODS: FOXD1 gene mutation screening, in 158 patients affected by PE, IUGR, RPL and repeated implantation failure (RIF), by direct sequencing and bioinformatics analysis. Plasmid constructs including FOXD1 mutations were used to perform in vitro gene reporter assays. RESULTS: Nine non-synonymous sequence variants were identified. Functional experiments revealed that p.His267Tyr and p.Arg57del led to disturbances of promoter transcriptional activity (C3 and PlGF genes). The FOXD1 p.Ala356Gly and p.Ile364Met deleterious mutations (previously found in RPL patients) have been identified in the present work in women suffering PE and IUGR. CONCLUSIONS: Our results argue in favour of FOXD1 mutations' central role in RPL, RIF, IUGR and PE pathogenesis via C3 and PlGF regulation and they describe, for the first time, a functional link between FOXD1 and implantation/placental diseases. FOXD1 could therefore be used in clinical environments as a molecular biomarker for these diseases in the near future.


Asunto(s)
Aborto Habitual/genética , Retardo del Crecimiento Fetal/genética , Factores de Transcripción Forkhead/genética , Predisposición Genética a la Enfermedad/genética , Preeclampsia/genética , Estudios de Cohortes , Femenino , Factores de Transcripción Forkhead/metabolismo , Humanos , Mutación/genética , Polimorfismo de Nucleótido Simple/genética , Embarazo , Regiones Promotoras Genéticas/genética
8.
Eur J Hum Genet ; 27(1): 49-60, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30206355

RESUMEN

We identified, through a genome-wide search for new imprinted genes in the human placenta, DSCAM (Down Syndrome Cellular Adhesion Molecule) as a paternally expressed imprinted gene. Our work revealed the presence of a Differentially Methylated Region (DMR), located within intron 1 that might regulate the imprinting in the region. This DMR showed a maternal allele methylation, compatible with its paternal expression. We showed that DSCAM is present in endothelial cells and the syncytiotrophoblast layer of the human placenta. In mouse, Dscam expression is biallelic in foetal brain and placenta excluding any possible imprinting in these tissues. This gene encodes a cellular adhesion molecule mainly known for its role in neurone development but its function in the placenta remains unclear. We report here the first imprinted gene located on human chromosome 21 with potential clinical implications.


Asunto(s)
Moléculas de Adhesión Celular/genética , Cromosomas Humanos Par 21/genética , Impresión Genómica , Placenta/metabolismo , Animales , Células COS , Células Cultivadas , Chlorocebus aethiops , Metilación de ADN , Femenino , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Humanos , Ratones , Ratones Endogámicos C57BL , Embarazo
10.
Sci Rep ; 6: 19196, 2016 Jan 13.
Artículo en Inglés | MEDLINE | ID: mdl-26758611

RESUMEN

Preeclampsia is a disease of pregnancy involving systemic endothelial dysfunction. However, cardiovascular consequences of preeclampsia are difficult to analyze in humans. The objective of the present study is to evaluate the cardiovascular dysfunction induced by preeclampsia by examining the endothelium of mice suffering of severe preeclampsia induced by STOX1 overexpression. Using Next Generation Sequencing on endothelial cells of mice carrying either transgenic or control embryos, we discovered significant alterations of gene networks involved in inflammation, cell cycle, and cardiac hypertrophy. In addition, the heart of the preeclamptic mice revealed cardiac hypertrophy associated with histological anomalies. Bioinformatics comparison of the networks of modified genes in the endothelial cells of the preeclamptic mice and HUVECs exposed to plasma from preeclamptic women identified striking similarities. The cardiovascular alterations in the pregnant mice are comparable to those endured by the cardiovascular system of preeclamptic women. The STOX1 mice could help to better understand the endothelial dysfunction in the context of preeclampsia, and guide the search for efficient therapies able to protect the maternal endothelium during the disease and its aftermath.


Asunto(s)
Cardiomegalia/etiología , Proteínas Portadoras/genética , Células Endoteliales/metabolismo , Preeclampsia/genética , Preeclampsia/metabolismo , Animales , Cardiomegalia/patología , Proteínas Portadoras/metabolismo , Línea Celular , Análisis por Conglomerados , Modelos Animales de Enfermedad , Femenino , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Redes Reguladoras de Genes , Humanos , Ratones , Ratones Transgénicos , Preeclampsia/mortalidad , Embarazo , Unión Proteica , Mapeo de Interacción de Proteínas , Mapas de Interacción de Proteínas , Transcriptoma
11.
Pediatr Res ; 77(6): 799-807, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25734244

RESUMEN

BACKGROUND: Intrauterine growth restriction (IUGR) is a frequent complication of pregnancy defined as a restriction of fetal growth. The objective of this work was to improve the knowledge on the pathophysiology of IUGR using a genome-wide method of expression analysis. METHODS: We analyzed differentially expressed genes in pooled placental tissues from vascular IUGR (four pools of three placentas) and normal pregnancies (four pools of three placentas) using a long nucleotide microarray platform (Nimblegen). We first did a global bioinformatics analysis based only on P value without any a priori. We secondly focused on "target" genes among the most modified ones. Finally, reverse transcription quantitative polymerase chain reaction (RT-qPCR) was performed on an extended panel of tissue samples (n = 62) on selected "target". RESULTS: We identified 636 modified genes among which 206 were upregulated (1.5 and higher; P < 0.05). Groups of patients were classified unambiguously. Genes involved in mitochondrial function and oxidative phosphorylation were decreased affecting three out of five complexes of the respiratory chain of the mitochondria, and thus energy production and metabolism. Among the most induced genes, we identified LEP, IGFBP1, and RBP4. CONCLUSION: Complementary studies on the role and function of LEP, IGFBP1, and RBP4 in IUGR pathophysiology and also in fetal programming remain necessary.


Asunto(s)
Retardo del Crecimiento Fetal/genética , Retardo del Crecimiento Fetal/fisiopatología , Regulación de la Expresión Génica/genética , Biología Computacional , Femenino , Perfilación de la Expresión Génica , Ontología de Genes , Humanos , Análisis por Micromatrices , Placenta/metabolismo , Embarazo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
12.
Antioxid Redox Signal ; 21(6): 819-34, 2014 Aug 20.
Artículo en Inglés | MEDLINE | ID: mdl-24738702

RESUMEN

AIMS: Storkhead box 1 (STOX1) is a winged-helix transcription factor that is implicated in the genetic forms of a high-prevalence human gestational disease, pre-eclampsia. STOX1 overexpression confers pre-eclampsia-like transcriptomic features to trophoblastic cell lines and pre-eclampsia symptoms to pregnant mice. The aim of this work was to evaluate the impact of STOX1 on free radical equilibrium and mitochondrial function, both in vitro and in vivo. RESULTS: Transcriptome analysis of STOX1-transgenic versus nontransgenic placentas at 16.5 days of gestation revealed alterations of mitochondria-related pathways. Placentas overexpressing STOX1 displayed altered mitochondrial mass and were severely biased toward protein nitration, indicating nitroso-redox imbalance in vivo. Trophoblast cells overexpressing STOX1 displayed an increased mitochondrial activity at 20% O2 and in hypoxia, despite reduction of the mitochondrial mass in the former. STOX1 overexpression is, therefore, associated with hyperactive mitochondria, resulting in increased free radical production. Moreover, nitric oxide (NO) production pathways were activated, resulting in peroxynitrite formation. At low oxygen pressure, STOX1 overexpression switched the free radical balance from reactive oxygen species (ROS) to reactive nitrogen species (RNS) in the placenta as well as in a trophoblast cell line. INNOVATION: In pre-eclamptic placentas, NO interacts with ROS and generates peroxynitrite and nitrated proteins as end products. This process will deprive the maternal organism of NO, a crucial vasodilator molecule. CONCLUSION: Our data posit STOX1 as a genetic switch in the ROS/RNS balance and suggest an explanation for elevated blood pressure in pre-eclampsia.


Asunto(s)
Proteínas Portadoras/genética , Homeostasis/genética , Mitocondrias/genética , Mitocondrias/metabolismo , Óxido Nítrico/metabolismo , Preeclampsia/genética , Preeclampsia/metabolismo , Animales , Modelos Animales de Enfermedad , Metabolismo Energético , Femenino , Regulación de la Expresión Génica , Hipoxia , Masculino , Ratones , Oxidación-Reducción , Placenta/metabolismo , Embarazo
13.
Genes Dev ; 28(5): 463-78, 2014 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-24589776

RESUMEN

Many loci maintain parent-of-origin DNA methylation only briefly after fertilization during mammalian development: Whether this form of transient genomic imprinting can impact the early embryonic transcriptome or even have life-long consequences on genome regulation and possibly phenotypes is currently unknown. Here, we report a maternal germline differentially methylated region (DMR) at the mouse Gpr1/Zdbf2 (DBF-type zinc finger-containing protein 2) locus, which controls the paternal-specific expression of long isoforms of Zdbf2 (Liz) in the early embryo. This DMR loses parental specificity by gain of DNA methylation at implantation in the embryo but is maintained in extraembryonic tissues. As a consequence of this transient, tissue-specific maternal imprinting, Liz expression is restricted to the pluripotent embryo, extraembryonic tissues, and pluripotent male germ cells. We found that Liz potentially functions as both Zdbf2-coding RNA and cis-regulatory RNA. Importantly, Liz-mediated events allow a switch from maternal to paternal imprinted DNA methylation and from Liz to canonical Zdbf2 promoter use during embryonic differentiation, which are stably maintained through somatic life and conserved in humans. The Gpr1/Zdbf2 locus lacks classical imprinting histone modifications, but analysis of mutant embryonic stem cells reveals fine-tuned regulation of Zdbf2 dosage through DNA and H3K27 methylation interplay. Together, our work underlines the developmental and evolutionary need to ensure proper Liz/Zdbf2 dosage as a driving force for dynamic genomic imprinting at the Gpr1/Zdbf2 locus.


Asunto(s)
Metilación de ADN , Impresión Genómica/genética , Mamíferos/genética , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo , Animales , Células Madre Embrionarias/metabolismo , Evolución Molecular , Femenino , Regulación del Desarrollo de la Expresión Génica , Histonas/metabolismo , Humanos , Masculino , Mamíferos/embriología , Mamíferos/metabolismo , Ratones , Regiones Promotoras Genéticas , Espermatogénesis/genética
14.
Epigenetics ; 9(1): 142-51, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24081307

RESUMEN

Preeclampsia (PE) is the major pregnancy-induced hypertensive disorder responsible for maternal and fetal morbidity and mortality that can be associated with intrauterine growth restriction (IUGR). PE and IUGR are thought to be due to a placental defect, occurring early during pregnancy. Several placental microRNAs (miRNAs) have been shown to be deregulated in the context of placental diseases and could thus play a role in the pathophysiology of PE. Here, we show that pri-miR-34a is overexpressed in preeclamptic placentas and that its placental expression is much higher during the first trimester of pregnancy than at term, suggesting a possible developmental role. We explored pri-miR-34a regulation and showed that P53, a known activator of miR-34a, is reduced in all pathological placentas and that hypoxia can induce pri-miR-34a expression in JEG-3 cells. We also studied the methylation status of the miR-34a promoter and revealed hypomethylation in all preeclamptic placentas (associated or not with IUGR), whereas hypoxia induced a hypermethylation in JEG-3 cells at 72 h. Despite the overexpression of pri-miR-34a in preeclampsia, there was a striking decrease of the mature miR-34a in this condition, suggesting preeclampsia-driven alteration of pri-miR-34a maturation. SERPINA3, a protease inhibitor involved in placental diseases, is elevated in IUGR and PE. We show here that miR-34a overexpression in JEG-3 downregulates SERPINA3. The low level of mature miR-34a could thus be an important mechanism contributing to SERPINA3 upregulation in placental diseases. Overall, our results support a role for miR-34a in the pathophysiology of preeclampsia, through deregulation of the pri-miRNA expression and its altered maturation.


Asunto(s)
Epigénesis Genética , MicroARNs/genética , MicroARNs/metabolismo , Enfermedades Placentarias/metabolismo , Línea Celular Tumoral , Coriocarcinoma/genética , Coriocarcinoma/metabolismo , Metilación de ADN , Femenino , Humanos , Hipoxia/metabolismo , Enfermedades Placentarias/genética , Preeclampsia/genética , Preeclampsia/metabolismo , Embarazo , Regiones Promotoras Genéticas , Serpinas/genética , Serpinas/metabolismo , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo , Neoplasias Uterinas/genética , Neoplasias Uterinas/metabolismo
15.
Front Genet ; 4: 248, 2013 Nov 21.
Artículo en Inglés | MEDLINE | ID: mdl-24312123

RESUMEN

MicroRNAs (miRNAs) have recently become essential actors in various fields of physiology and medicine, especially as easily accessible circulating biomarkers, or as modulators of cell differentiation. To this respect, terminal differentiation of trophoblasts (the characteristic cells of the placenta in Therian mammals) into syncytiotrophoblast, villous trophoblast, or extravillous trophoblast constitutes a good example of such a choice, where miRNAs have recently been shown to play an important role. The aim of this review is to provide a snapshot of what is known today in placentation mechanisms that are mediated by miRNA, under the angles of materno-fetal immune dialog regulation, trophoblast differentiation, and angiogenesis at the materno-fetal interface. Also, two aspects of regulation of these issues will be highlighted: the part played by oxygen concentration and the specific function of imprinted genes in the developing placenta.

16.
Hypertension ; 61(3): 662-8, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23357179

RESUMEN

Preeclampsia (PE) is a common human-specific pregnancy disorder defined by hypertension and proteinuria during gestation and responsible for maternal and fetal morbimortality. STOX1, encoding a transcription factor, was the first gene associated with PE as identified by positional cloning approaches. Its overexpression in choriocarcinoma cells mimics the transcriptional consequences of PE in the human placenta. Here, we created transgenic mouse strains overexpressing human STOX1. Wild-type female mice crossed with transgenic male mice reproduce accurately the symptoms of severe PE: gestational hypertension, proteinuria, and elevated plasma levels of soluble fms-like tyrosine kinase 1 and soluble endoglin. Placental and kidney histology were altered. Symptoms were prevented or alleviated by aspirin treatment. STOX1-overexpressing mice constitute a unique model for studying PE, allow testing therapeutic approaches, and assessing the long-term effects of the preeclamptic syndrome.


Asunto(s)
Antiinflamatorios no Esteroideos/uso terapéutico , Antihipertensivos/uso terapéutico , Aspirina/uso terapéutico , Proteínas Portadoras/biosíntesis , Placenta/metabolismo , Preeclampsia/tratamiento farmacológico , Animales , Proteínas Portadoras/efectos adversos , Proteínas Portadoras/genética , Modelos Animales de Enfermedad , Endoglina , Femenino , Humanos , Péptidos y Proteínas de Señalización Intracelular/sangre , Riñón/patología , Masculino , Ratones , Ratones Transgénicos , Placenta/patología , Preeclampsia/etiología , Preeclampsia/genética , Embarazo , Índice de Severidad de la Enfermedad , Receptor 1 de Factores de Crecimiento Endotelial Vascular/sangre
17.
Epigenetics ; 7(9): 1079-90, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22894909

RESUMEN

Genomic imprinting characterizes genes with a monoallelic expression, which is dependent on the parental origin of each allele. Approximately 150 imprinted genes are known to date, in humans and mice but, though computational searches have tried to extract intrinsic characteristics of these genes to identify new ones, the existing list is probably far from being comprehensive. We used a high-throughput strategy by diverting the classical use of genotyping microarrays to compare the genotypes of mRNA/cDNA vs. genomic DNA to identify new genes presenting monoallelic expression, starting from human placental material. After filtering of data, we obtained a list of 1,082 putative candidate monoallelic SNPs located in more than one hundred candidate genes. Among these, we found known imprinted genes, such as IPW, GRB10, INPP5F and ZNF597, which contribute to validate the approach. We also explored some likely candidates of our list and identified seven new imprinted genes, including ZFAT, ZFAT-AS1, GLIS3, NTM, MAGI2, ZC3H12Cand LIN28B, four of which encode zinc finger transcription factors. They are, however, not imprinted in the mouse placenta, except for Magi2. We analyzed in more details the ZFAT gene, which is paternally expressed in the placenta (as ZFAT-AS1, a non-coding antisense RNA) but biallelic in other tissues. The ZFAT protein is expressed in endothelial cells, as well as in syncytiotrophoblasts. The expression of this gene is, moreover, downregulated in placentas from complicated pregnancies. With this work we increase by about 10% the number of known imprinted genes in humans.


Asunto(s)
Genoma Humano/genética , Impresión Genómica/genética , Placenta/metabolismo , Animales , Bovinos , Femenino , Perfilación de la Expresión Génica , Sitios Genéticos , Humanos , Ratones , Ratones Endogámicos C57BL , Análisis de Secuencia por Matrices de Oligonucleótidos , Polimorfismo de Nucleótido Simple , Embarazo , ARN Mensajero/biosíntesis , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
18.
Hum Mol Genet ; 21(9): 1968-78, 2012 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-22246292

RESUMEN

SERPINA3 (Serpin peptidase inhibitor clade A member 3), also known as a1-antichymotrypsin, is a serine protease inhibitor involved in a wide range of biological processes. Recently, it has been shown to be up-regulated in human placental diseases in association with a hypomethylation of the 5' region of the gene. In the present study, we show that the promoter of SERPINA3 is transcriptionally activated by three transcription factors (TFs) (SP1, MZF1 and ZBTB7B), the level of induction being dependent on the rs1884082 single nucleotide polymorphism (SNP) located inside the promoter, the T allele being consistently induced to a higher level than the G, with or without added TFs. When the promoter was methylated, the response to ZBTB7B was allele specific (the G allele was strongly induced, while the T allele was strongly down-regulated). We propose an adaptive model to explain the interest of such a regulation for placental function and homeostasis. Overexpression of SERPINA3 in JEG-3 cells, a trophoblast cell model, decreased cell adhesion to the extracellular matrix and to neighboring cells, but protects them from apoptosis, suggesting a way by which this factor could be deleterious at high doses. In addition, we show in different human populations that the T allele appears to predispose to Intra Uterine Growth Restriction (IUGR), while a G allele at a second SNP located in the second exon (rs4634) increases the risk of preeclampsia. Our results provide mechanistic views inside the involvement of SERPINA3 in placental diseases, through its regulation by a combination of epigenetic, genetic and TF-mediated regulations.


Asunto(s)
Enfermedades Placentarias/genética , Serpinas/genética , Alelos , Apoptosis , Secuencia de Bases , Estudios de Casos y Controles , Adhesión Celular , Línea Celular , Metilación de ADN , Proteínas de Unión al ADN/metabolismo , Epigénesis Genética , Femenino , Retardo del Crecimiento Fetal/genética , Retardo del Crecimiento Fetal/metabolismo , Regulación de la Expresión Génica , Estudios de Asociación Genética , Predisposición Genética a la Enfermedad , Humanos , Hipoxia/genética , Hipoxia/metabolismo , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Factores de Transcripción de Tipo Kruppel/metabolismo , Modelos Biológicos , Enfermedades Placentarias/metabolismo , Polimorfismo de Nucleótido Simple , Preeclampsia/genética , Preeclampsia/metabolismo , Embarazo , Regiones Promotoras Genéticas , ARN Mensajero/genética , ARN Mensajero/metabolismo , Factor de Transcripción Sp1/metabolismo , Factores de Transcripción/metabolismo , Activación Transcripcional , Trofoblastos/citología , Trofoblastos/metabolismo , Dedos de Zinc
19.
PLoS One ; 6(6): e21222, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21731679

RESUMEN

A low-protein diet applied during pregnancy in the rat results in intrauterine growth restricted (IUGR) fetuses. In humans, IUGR is associated with increased perinatal morbidity, higher incidence of neuro-developmental defects and increased risk of adult metabolic anomalies, such as diabetes and cardiovascular disease. Development and function of many organs are affected by environmental conditions such as those inducing fetal and early postnatal growth restriction. This phenomenon, termed "fetal programming" has been studied unconnectedly in some organs, but very few studies (if any) have investigated at the same time several organs, on a more comparative basis. However, it is quite probable that IUGR affects differentially most organ systems, with possible persistent changes in gene expression. In this study we address transcriptional alterations induced by IUGR in a multi-organ perspective, by systematic analysis of 20-days rat fetuses. We show that (1) expressional alterations are apparently stronger in organs functioning late in foetal or postnatal life than in organs that are functioning early (2) hierarchical classification of the deregulations put together kidney and placenta in one cluster, liver, lungs and heart in another; (3) the epigenetic machinery is set up especially in the placenta, while its alterations are rather mild in other organs; (4) the genes appear deregulated in chromosome clusters; (5) the altered expression cascades varies from organ to organ, with noticeably a very significant modification of the complement and coagulation cascades in the kidney; (6) we found a significant increase in TF binding site for HNF4 proteins specifically for liver genes that are down-regulated in IUGR, suggesting that this decrease is achieved through the action of HNF transcription factors, that are themselves transcriptionnally induced in the liver by IUGR (x 1.84 fold). Altogether, our study suggests that a combination of tissue-specific mechanisms contributes to bring about tissue-driven modifications of gene cascades. The question of these cascades being activated to adapt the organ to harsh environmental condition, or as an endpoint consequence is still raised.


Asunto(s)
Retardo del Crecimiento Fetal/genética , Retardo del Crecimiento Fetal/fisiopatología , Perfilación de la Expresión Génica , Regulación del Desarrollo de la Expresión Génica , Especificidad de Órganos/genética , Animales , Cromosomas de los Mamíferos/genética , Análisis por Conglomerados , Modelos Animales de Enfermedad , Regulación hacia Abajo/genética , Epigénesis Genética , Femenino , Impresión Genómica/genética , Embarazo , Regiones Promotoras Genéticas/genética , Ratas , Ratas Sprague-Dawley , Transducción de Señal/genética , Estrés Fisiológico/genética
20.
Epigenetics ; 6(2): 247-55, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-20962579

RESUMEN

Preeclampsia (PE) and vascular intra-uterine growth restriction (vIUGR) are two pathological obstetrical conditions originating from placental dysfunction. Recently, methylation changes at the placental level have been shown to be indicative of these diseases. The alteration of such epigenetic marks is therefore a novel pathway that might be critical for these pathologies. Here, we identified a region located in the distal promoter of the T-box-containing transcription factor TBX15 that is differentially methylated in pathological placentas. The level of methylation correlated significantly with the weight and stature of the newborn. The promoter was found to be hypomethylated in vIUGR coinciding with the down-regulation of its expression. PDX1, a transcription factor important for the regulation of insulin metabolism regulation was able to repress the TBX15 promoter in a methylation-dependent manner, which might, at least partially, explain the specific mRNA decrease of TBX15 observed in vIUGR placentas. Overall, the data presented herein suggest that TBX15 might be involved in the pathophysiology of placental diseases.


Asunto(s)
Metilación de ADN/genética , Retardo del Crecimiento Fetal/genética , Proteínas de Homeodominio/genética , Enfermedades Placentarias/genética , Preeclampsia/genética , Proteínas de Dominio T Box/genética , Transactivadores/genética , Adulto , Secuencia de Bases , Línea Celular , Epigenómica , Femenino , Expresión Génica/genética , Humanos , Recién Nacido , Datos de Secuencia Molecular , Placenta/metabolismo , Embarazo , Regiones Promotoras Genéticas/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...