Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 20
Filtrar
1.
Am J Obstet Gynecol ; 228(4): 409-417.e4, 2023 04.
Artigo em Inglês | MEDLINE | ID: mdl-36209938

RESUMO

OBJECTIVE: This study aimed to determine the incremental yield of prenatal exome sequencing over chromosomal microarray or G-banding karyotype in fetuses with: (1) intrauterine growth restriction related to placental insufficiency or (2) short long bones, in isolated and nonisolated instances for both scenarios. DATA SOURCES: Data were collected via electronic searches for relevant citations from January 2010 to April 10, 2022 in MEDLINE, Embase, Web of Science, and Cochrane, and using relevant bibliographies and data generated in-house. STUDY ELIGIBILITY CRITERIA: Included were prospective or retrospective cohort studies and/or case series with: (1) n>5 cases of short long bones and/or intrauterine growth restriction undergoing prenatal sequencing with a clearly defined phenotype including assessment of placental function; (2) testing based on prenatal phenotype only; (3) a nondiagnostic chromosomal microarray/karyotype; and (4) known results of genetic testing. METHODS: Incremental yield was calculated for each study and as a pooled value for the aforementioned groups using a random-effects model. Results were displayed in forest plots with 95% confidence intervals. Heterogeneity was assessed statistically using Higgins' I2. Publication bias was assessed graphically using funnel plots. Quality assessment was performed using modified Standards for Reporting of Diagnostic Accuracy criteria (International Prospective Register of Systematic Reviews registration number CRD42022324680). RESULTS: Nineteen studies were included (n=452 cases). The apparent incremental yields with prenatal sequencing were: (1) 4% (95% confidence interval, -5.0 to 12; I2=0%) in isolated intrauterine growth restriction with evidence of placental insufficiency, (2) 30% (95% confidence interval, 13-47; I2=1%) in intrauterine growth restriction with additional structural anomalies, (3) 48% (95% confidence interval, 26-70; I2=73%) in isolated short long bones, and (4) 68% (95% confidence interval, 58-77; I2=51%) in short long bones with additional skeletal anomalies. Of the 37 short long bone cases with a diagnosis, 32 had a skeletal dysplasia, with thanatophoric dysplasia and osteogenesis imperfecta being the most common (both 21.6% [n=8/37]). In fetuses with short long bones and additional skeletal features, osteogenesis imperfecta was the most common diagnosis (28% [n=57/204]). Where documented, the inheritance patterns were de novo in 75.4% (n=150) of cases. CONCLUSION: Prenatal sequencing adds substantially to incremental yield over chromosomal microarray in fetuses with short long bones or multisystem intrauterine growth restriction. Robust studies are required to assess the utility of fetal sequencing in isolated intrauterine growth restriction with evidence of placental insufficiency, which cannot be recommended on the basis of current evidence.


Assuntos
Osteogênese Imperfeita , Insuficiência Placentária , Humanos , Gravidez , Feminino , Retardo do Crescimento Fetal/diagnóstico , Retardo do Crescimento Fetal/genética , Insuficiência Placentária/genética , Sequenciamento do Exoma , Estudos Retrospectivos , Placenta , Diagnóstico Pré-Natal/métodos , Ultrassonografia Pré-Natal
2.
BMC Pregnancy Childbirth ; 22(1): 77, 2022 Jan 28.
Artigo em Inglês | MEDLINE | ID: mdl-35090410

RESUMO

BACKGROUND: Intrauterine growth restriction (IUGR) is defined as a fetus that fails to achieve its genetically determined growth potential. The exact molecular mechanisms of placental insufficiency IUGR pathogenesis are a little known. Our goal was to identify key genes and gene co-expression modules related to placental insufficiency IUGR. METHODS: We used weighted gene co-expression network analysis (WGCNA) and protein-protein interaction (PPI) network analysis to examine the IUGR dataset GSE114691 from NCBI Gene Expression Omnibus. Core modules and hub nodes of the protein-protein interaction network were identified. A gene network was constructed and genes were classified by WGCNA into different modules. The validation of potential key genes was carried out using additional datasets (GSE12216 and GSE24129). RESULTS: We identified in GSE114691 539 down regulated genes and 751 up regulated genes in placental tissues characteristic of placental insufficiency IUGR compared with non-IUGR, and defined 76 genes as hub nodes in the protein-protein interaction network. Genes in the key modules of the WGCNA network were most closely associated with placental insufficiency IUGR and significantly enriched in biological process such as cellular metabolic process and macromolecule metabolic process. We identified as key genes TGFB1, LEP, ENG, ITGA5, STAT5A, LYN, GATA3, FPR1, TGFB2, CEBPB, KLF4, FLT1, and PNPLA2. The RNA expression levels of ENG and LEP, as biomarkers, were validated. CONCLUSION: A holistic gene expression profile of placental insufficiency IUGR has been generated and the key genes ENG and LEP has potential to serve as circulating diagnosis biomarkers and therapeutic targets for placental insufficiency IUGR.


Assuntos
Endoglina/genética , Retardo do Crescimento Fetal/genética , Perfilação da Expressão Gênica , Redes Reguladoras de Genes , Leptina/genética , Insuficiência Placentária/genética , Mapas de Interação de Proteínas , Biomarcadores , Bases de Dados Genéticas , Regulação para Baixo , Feminino , Humanos , Gravidez , Reprodutibilidade dos Testes , Regulação para Cima , Receptor 1 de Fatores de Crescimento do Endotélio Vascular/genética
3.
Hum Gene Ther ; 31(21-22): 1190-1202, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-32988220

RESUMO

Severe fetal growth restriction (FGR) affects 1:500 pregnancies, is untreatable and causes serious neonatal morbidity and death. Reduced uterine blood flow (UBF) and lack of bioavailable VEGF due to placental insufficiency is a major cause. Transduction of uterine arteries in normal or FGR sheep and guinea pigs using an adenovirus (Ad) encoding VEGF isoforms A (Ad.VEGF-A165) and a FLAG-tagged pre-processed short form D (DΔNΔC, Ad.VEGF-DΔNΔC-FLAG) increases endothelial nitric oxide expression, enhances relaxation and reduces constriction of the uterine arteries and their branches. UBF and angiogenesis are increased long term, improving fetal growth in utero. For clinical trial development we compared Ad.VEGF vector transduction efficiency and function in endothelial cells (ECs) derived from different species. We aimed to compare the transduction efficiency and function of the pre-clinical study Ad. constructs (Ad.VEGF-A165, Ad.VEGF-DΔNΔC-FLAG) with the intended clinical trial construct (Ad.VEGF-DΔNΔC) where the FLAG tag is removed. We infected ECs from human umbilical vein, pregnant sheep uterine artery, pregnant guinea pig aorta and non-pregnant rabbit aorta, with increasing multiplicity of infection (MOI) for 24 or 48 hours of three Ad.VEGF vectors, compared to control Ad. containing the LacZ gene (Ad.LacZ). VEGF supernatant expression was analysed by ELISA. Functional assessment used tube formation assay and Erk-Akt phosphorylation by ELISA. VEGF expression was higher after Ad.VEGF-DΔNΔC-FLAG and Ad.VEGF-DΔNΔC transduction compared to Ad.VEGF-A165 in all EC types (*p < 0.001). Tube formation was higher in ECs transduced with Ad.VEGF-DΔNΔC in all species compared to other constructs (***p < 0.001, *p < 0.05 with rabbit aortic ECs). Phospho-Erk and phospho-Akt assays displayed no differences between the three vector constructs, whose effect was, as in other experiments, higher than Ad.LacZ (***p < 0.001). In conclusion, we observed high transduction efficiency and functional effects of Ad.VEGF-DΔNΔC vector with comparability in major pathway activation to constructs used in pre-clinical studies, supporting its use in a clinical trial.


Assuntos
Adenoviridae/genética , Dependovirus/genética , Endotélio Vascular/metabolismo , Terapia Genética/métodos , Vetores Genéticos/administração & dosagem , Insuficiência Placentária/terapia , Fator A de Crescimento do Endotélio Vascular/genética , Animais , Feminino , Vetores Genéticos/genética , Cobaias , Células Endoteliais da Veia Umbilical Humana , Humanos , Insuficiência Placentária/genética , Insuficiência Placentária/metabolismo , Insuficiência Placentária/patologia , Gravidez , Coelhos , Ovinos
4.
Dev Dyn ; 249(2): 237-244, 2020 02.
Artigo em Inglês | MEDLINE | ID: mdl-31625212

RESUMO

Small ubiquitin-related modifier (SUMO)-specific protease 2 (SENP2) is essential for the development of healthy placenta. The loss of SENP2 causes severe placental deficiencies and leads to embryonic death that is associated with heart and brain deformities. However, tissue-specific disruption of SENP2 demonstrates its dispensable role in embryogenesis and the embryonic defects are secondary to placental insufficiency. SENP2 regulates SUMO1 modification of Mdm2, which controls p53 activities critical for trophoblast cell proliferation and differentiation. Here we use genetic analyses to examine the involvement of SUMO2 and SUMO3 for SENP2-mediated placentation. The results indicate that hyper-SUMOylation caused by SENP2 deficiency can be compensated by reducing the level of SUMO modifiers. The placental deficiencies caused by the loss of SENP2 can be alleviated by the inactivation of gene encoding SUMO2 or SUMO3. Our findings demonstrate that SENP2 genetically interacts with SUMO2 and SUMO3 pivotal for the development of three major trophoblast layers. The alleviation of placental defects in the SENP2 knockouts further leads to the proper formation of the heart structures, including atrioventricular cushion and myocardium. SUMO2 and SUMO3 modifications regulate placentation and organogenesis mediated by SENP2.


Assuntos
Cisteína Endopeptidases/metabolismo , Embrião de Mamíferos/metabolismo , Proteínas Modificadoras Pequenas Relacionadas à Ubiquitina/metabolismo , Ubiquitinas/metabolismo , Animais , Cisteína Endopeptidases/genética , Feminino , Camundongos , Camundongos Knockout , Insuficiência Placentária/genética , Insuficiência Placentária/metabolismo , Gravidez , Proteínas Modificadoras Pequenas Relacionadas à Ubiquitina/genética , Trofoblastos/metabolismo , Ubiquitinas/genética
5.
J Pathol ; 242(2): 246-259, 2017 06.
Artigo em Inglês | MEDLINE | ID: mdl-28295343

RESUMO

Epithelial membrane protein-2 (EMP2) is a tetraspan protein predicted to regulate placental development. Highly expressed in secretory endometrium and trophectoderm cells, previous studies suggest that it may regulate implantation by orchestrating the surface expression of integrins and other membrane proteins. In order to test the role of EMP2 in pregnancy, mice lacking EMP2 (Emp2-/- ) were generated. Emp2-/- females are fertile but have reduced litter sizes when carrying Emp2-/- but not Emp2+/- fetuses. Placentas of Emp2-/- fetuses exhibit dysregulation in pathways related to neoangiogenesis, coagulation, and oxidative stress, and have increased fibrin deposition and altered vasculature. Given that these findings often occur due to placental insufficiency resulting in an oxygen-poor environment, the expression of hypoxia-inducible factor-1 alpha (HIF-1α) was examined. Placentas from Emp2-/- fetuses had increased total HIF-1α expression in large part through an increase in uterine NK (uNK) cells, demonstrating a unique interplay between uNK cells and trophoblasts modulated through EMP2. To determine if these results translated to human pregnancy, placentas from normal, term deliveries or those complicated by placental insufficiency resulting in intrauterine growth restriction (IUGR) were stained for EMP2. EMP2 was significantly reduced in both villous and extravillous trophoblast populations in IUGR placentas. Experiments in vitro using human trophoblast cells lines indicate that EMP2 modulates angiogenesis by altering HIF-1α expression. Our results reveal a novel role for EMP2 in regulating trophoblast function and vascular development in mice and humans, and suggest that it may be a new biomarker for placental insufficiency. Copyright © 2017 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.


Assuntos
Retardo do Crescimento Fetal/genética , Glicoproteínas de Membrana/genética , Oxigênio/metabolismo , Insuficiência Placentária/genética , Animais , Modelos Animais de Doenças , Feminino , Retardo do Crescimento Fetal/metabolismo , Retardo do Crescimento Fetal/patologia , Fibrina/genética , Fibrina/metabolismo , Técnicas de Inativação de Genes , Recombinação Homóloga , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Masculino , Glicoproteínas de Membrana/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Neovascularização Patológica , Placenta/irrigação sanguínea , Placenta/metabolismo , Placenta/patologia , Insuficiência Placentária/metabolismo , Insuficiência Placentária/patologia , Placentação , Gravidez , Trofoblastos/metabolismo , Trofoblastos/patologia , Útero/irrigação sanguínea , Útero/metabolismo , Útero/patologia
6.
J Endocrinol ; 232(2): 247-257, 2017 02.
Artigo em Inglês | MEDLINE | ID: mdl-27885054

RESUMO

Prenatal events such as intrauterine growth restriction can affect gonadal development of the offspring and have an impact on reproductive health. To investigate the effects of intrauterine growth restriction induced by uterine artery ligation on the postnatal rat testis. Pregnant rats underwent uterine artery ligation at day 19 of gestation. Offspring were killed at 5, 20 and 40 days post-partum (dpp). At killing, one gonad was snap-frozen in liquid nitrogen and processed for RNA and steroid extraction. The other gonad was formalin-fixed for histology. Gene expression was analyzed by TaqMan Low-Density Array. Intratesticular testosterone, estradiol and serum gonadotrophins were measured. Thirty genes were dysregulated in intrauterine growth-restricted rats compared to controls, among which markers of Sertoli cell and Leydig cell function, cell metabolism and growth factors. Testis weights were significantly reduced at 5 and 20 dpp in intrauterine growth-restricted rats and caught-up by 40 dpp Accordingly, Sertoli cell number was significantly lower in 5 dpp intrauterine growth-restricted rats. At 20 dpp, intratesticular testosterone was significantly increased in intrauterine growth-restricted rats, whereas serum gonadotrophins were unchanged. IUGR altered the gene expression in the rat testes up to peripubertal age and reduced testis size and Sertoli cell number in neonatal age. Multiple mechanisms encompassing genetic changes and steroid production by the testis may be involved in the catch-up growth phase that restored testis size by 40 dpp Permanent consequences on organ function and gamete integrity cannot be excluded and deserve further investigations.


Assuntos
Retardo do Crescimento Fetal/metabolismo , Insuficiência Placentária/metabolismo , Testículo/metabolismo , Testosterona/metabolismo , Animais , Estradiol/metabolismo , Feminino , Retardo do Crescimento Fetal/genética , Hormônio Foliculoestimulante/sangue , Expressão Gênica , Células Intersticiais do Testículo/metabolismo , Hormônio Luteinizante/sangue , Masculino , Insuficiência Placentária/genética , Gravidez , Ratos , Ratos Sprague-Dawley , Células de Sertoli/metabolismo
7.
Endocrinology ; 157(5): 1813-25, 2016 05.
Artigo em Inglês | MEDLINE | ID: mdl-27007072

RESUMO

Leptin availability in perinatal life critically affects metabolic programming. We tested the hypothesis that uteroplacental insufficiency and intrauterine stress affect perinatal leptin availability in rat offspring. Pregnant rats underwent bilateral uterine vessel ligation (LIG; n = 14), sham operation (SOP; n = 12), or no operation (controls, n = 14). Fetal livers (n = 180), placentas (n = 180), and maternal blood were obtained 4 hours (gestational day [E] 19), 24 hours (E20), and 72 hours (E22) after surgery. In the offspring, we took blood samples on E22 (n = 44), postnatal day (P) 1 (n = 29), P2 (n = 16), P7 (n = 30), and P12 (n = 30). Circulating leptin (ELISA) was significantly reduced in LIG (E22, P1, P2) and SOP offspring (E22). Postnatal leptin surge was delayed in LIG but was accelerated in SOP offspring. Placental leptin gene expression (quantitative RT-PCR) was reduced in LIG (E19, E20, E22) and SOP (E20, E22). Hepatic leptin receptor (Lepr-a, mediating leptin degradation) gene expression was increased in LIG fetuses (E20, E22) only. Surprisingly, hypoxia-inducible factors (Hif; Western blot) were unaltered in placentas and were reduced in the livers of LIG (Hif1a, E20; Hif2a, E19, E22) and SOP (Hif2a, E19) fetuses. Gene expression of prolyl hydroxylase 3, a factor expressed under hypoxic conditions contributing to Hif degradation, was increased in livers of LIG (E19, E20, E22) and SOP (E19) fetuses and in placentas of LIG and SOP (E19). In summary, reduced placental leptin production, increased fetal leptin degradation, and persistent perinatal hypoleptinemia are present in intrauterine growth restriction offspring, especially after uteroplacental insufficiency, and may contribute to perinatal programming of leptin resistance and adiposity in later life.


Assuntos
Leptina/metabolismo , Placenta/metabolismo , Insuficiência Placentária/metabolismo , Receptores para Leptina/metabolismo , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Feminino , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Leptina/sangue , Leptina/genética , Fígado/metabolismo , Insuficiência Placentária/sangue , Insuficiência Placentária/genética , Gravidez , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Ratos , Ratos Wistar , Receptores para Leptina/genética
8.
PLoS One ; 10(10): e0140879, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26473479

RESUMO

Low birth weight is associated with both short term problems and the fetal programming of adult onset diseases, including an increased risk of obesity, diabetes and cardiovascular disease. Placental insufficiency leading to intrauterine growth restriction (IUGR) contributes to the prevalence of diseases with developmental origins. Currently there are no therapies for IUGR or placental insufficiency. To address this and move towards development of an in utero therapy, we employ a nanostructure delivery system complexed with the IGF-1 gene to treat the placenta. IGF-1 is a growth factor critical to achieving appropriate placental and fetal growth. Delivery of genes to a model of human trophoblast and mouse placenta was achieved using a diblock copolymer (pHPMA-b-pDMAEMA) complexed to hIGF-1 plasmid DNA under the control of trophoblast-specific promoters (Cyp19a or PLAC1). Transfection efficiency of pEGFP-C1-containing nanocarriers in BeWo cells and non-trophoblast cells was visually assessed via fluorescence microscopy. In vivo transfection and functionality was assessed by direct placental-injection into a mouse model of IUGR. Complexes formed using pHPMA-b-pDMAEMA and CYP19a-923 or PLAC1-modified plasmids induce trophoblast-selective transgene expression in vitro, and placental injection of PLAC1-hIGF-1 produces measurable RNA expression and alleviates IUGR in our mouse model, consequently representing innovative building blocks towards human placental gene therapies.


Assuntos
Técnicas de Transferência de Genes , Terapia Genética/métodos , Fator de Crescimento Insulin-Like I/biossíntese , Insuficiência Placentária/terapia , Trofoblastos/metabolismo , Animais , Aromatase/genética , Linhagem Celular Tumoral , Modelos Animais de Doenças , Feminino , Humanos , Fator de Crescimento Insulin-Like I/genética , Camundongos , Insuficiência Placentária/genética , Insuficiência Placentária/metabolismo , Insuficiência Placentária/patologia , Gravidez , Proteínas da Gravidez/genética , Regiões Promotoras Genéticas , Trofoblastos/patologia
9.
Hum Gene Ther ; 26(3): 172-82, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25738403

RESUMO

Intrauterine growth restriction (IUGR) due to placental insufficiency is a leading cause of perinatal complications for which there is no effective prenatal therapy. We have previously demonstrated that intraplacental injection of adenovirus-mediated insulin-like growth factor-1 (Ad-IGF-1) corrects fetal weight in a murine IUGR model induced by mesenteric uterine artery branch ligation. This study investigated the effect of intraplacental Ad-IGF-1 gene therapy in a rabbit model of naturally occurring IUGR (runt) due to placental insufficiency, which is similar to the human IUGR condition with onset in the early third trimester, brain sparing, and a reduction in liver weight. Laparotomy was performed on New Zealand White rabbits on day 21 of 30 days of gestation and litters were divided into five groups: Control (first position)+phosphate-buffered saline (PBS), control+Ad-IGF-1, runt (third position)+PBS, runt+Ad-IGF-1, and runt+Ad-LacZ. The effect of IGF-1 gene therapy on fetal, placental, liver, heart, lung, and musculoskeletal weights of the growth-restricted pups was examined. Protein expression after gene transfer was seen along the maternal-fetal placenta interface (n=12) 48 hr after gene therapy. There was minimal gene transfer detected in the pups or maternal organs. At term, compared with the normally grown first-position control, the runted third-position pups demonstrated significantly lower fetal, placental, liver, lung, and musculoskeletal weights. The fetal, liver, and musculoskeletal weights were restored to normal by intraplacental Ad-IGF-1 gene therapy (p<0.01), with no change in the placental weight. Intraplacental gene therapy is a novel strategy for the treatment of IUGR caused by placental insufficiency that takes advantage of an organ that will be discarded at birth. Development of nonviral IGF-1 gene delivery using placenta-specific promoters can potentially minimize toxicity to the mother and fetus and facilitate clinical translation of this novel therapy.


Assuntos
Retardo do Crescimento Fetal/genética , Retardo do Crescimento Fetal/terapia , Terapia Genética/métodos , Fator de Crescimento Insulin-Like I/farmacologia , Placenta/metabolismo , Insuficiência Placentária/genética , Adenoviridae , Animais , Feminino , Feto/efeitos dos fármacos , Técnicas de Transferência de Genes , Coração/efeitos dos fármacos , Coração/crescimento & desenvolvimento , Fator de Crescimento Insulin-Like I/administração & dosagem , Fator de Crescimento Insulin-Like I/genética , Fígado/efeitos dos fármacos , Fígado/crescimento & desenvolvimento , Pulmão/efeitos dos fármacos , Pulmão/crescimento & desenvolvimento , Músculo Esquelético/efeitos dos fármacos , Músculo Esquelético/crescimento & desenvolvimento , Tamanho do Órgão/efeitos dos fármacos , Placenta/efeitos dos fármacos , Gravidez , Coelhos
10.
J Am Heart Assoc ; 3(4)2014 Jul 28.
Artigo em Inglês | MEDLINE | ID: mdl-25085511

RESUMO

BACKGROUND: Placental insufficiency is the leading cause of intrauterine growth restriction in the developed world and results in chronic hypoxemia in the fetus. Oxygen is essential for fetal heart development, but a hypoxemic environment in utero can permanently alter development of cardiomyocytes. The present study aimed to investigate the effect of placental restriction and chronic hypoxemia on total number of cardiomyocytes, cardiomyocyte apoptosis, total length of coronary capillaries, and expression of genes regulated by hypoxia. METHODS AND RESULTS: We induced experimental placental restriction from conception, which resulted in fetal growth restriction and chronic hypoxemia. Fetal hearts in the placental restriction group had fewer cardiomyocytes, but interestingly, there was no difference in the percentage of apoptotic cardiomyocytes; the abundance of the transcription factor that mediates hypoxia-induced apoptosis, p53; or expression of apoptotic genes Bax and Bcl2. Likewise, there was no difference in the abundance of autophagy regulator beclin 1 or expression of autophagic genes BECN1, BNIP3, LAMP1, and MAP1LC3B. Furthermore, fetuses exposed to normoxemia (control) or chronic hypoxemia (placental restriction) had similar mRNA expression of a suite of hypoxia-inducible factor target genes, which are essential for angiogenesis (VEGF, Flt1, Ang1, Ang2, and Tie2), vasodilation (iNOS and Adm), and glycolysis (GLUT1 and GLUT3). In addition, there was no change in the expression of PKC-ε, a cardioprotective gene with transcription regulated by hypoxia in a manner independent of hypoxia-inducible factors. There was an increased capillary length density but no difference in the total length of capillaries in the hearts of the chronically hypoxemic fetuses. CONCLUSION: The lack of upregulation of hypoxia target genes in response to chronic hypoxemia in the fetal heart in late gestation may be due to a decrease in the number of cardiomyocytes (decreased oxygen demand) and the maintenance of the total length of capillaries. Consequently, these adaptive responses in the fetal heart may maintain a normal oxygen tension within the cardiomyocyte of the chronically hypoxemic fetus in late gestation.


Assuntos
Regulação da Expressão Gênica no Desenvolvimento , Hipóxia/genética , Miócitos Cardíacos/metabolismo , Insuficiência Placentária/genética , RNA Mensageiro/metabolismo , Animais , Apoptose/genética , Proteínas Reguladoras de Apoptose/genética , Proteínas Reguladoras de Apoptose/metabolismo , Contagem de Células , Feminino , Genes bcl-2/genética , Transportador de Glucose Tipo 1/genética , Transportador de Glucose Tipo 1/metabolismo , Transportador de Glucose Tipo 3/genética , Transportador de Glucose Tipo 3/metabolismo , Hipóxia/metabolismo , Proteína 1 de Membrana Associada ao Lisossomo/genética , Proteína 1 de Membrana Associada ao Lisossomo/metabolismo , Miócitos Cardíacos/citologia , Óxido Nítrico Sintase Tipo II/genética , Óxido Nítrico Sintase Tipo II/metabolismo , Insuficiência Placentária/metabolismo , Gravidez , Ovinos , Proteína Supressora de Tumor p53/genética , Proteína Supressora de Tumor p53/metabolismo , Fator A de Crescimento do Endotélio Vascular/genética , Fator A de Crescimento do Endotélio Vascular/metabolismo , Proteína X Associada a bcl-2/genética , Proteína X Associada a bcl-2/metabolismo
11.
Curr Opin Endocrinol Diabetes Obes ; 21(3): 227-32, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24722170

RESUMO

PURPOSE OF REVIEW: The purpose of this study was to review and comment on recent original presentations dealing with genetic and environmental factors in the cause of hypospadias. RECENT FINDINGS: The heritability is definitely high and having an affected family member is the highest identified risk factor so far. Many candidate genes and polymorphisms have been suggested for hypospadias. Some associations with hypospadias were found, and many of these were replicated inconsistently as would be expected in a complex disorder affected by both genes and environment. The consistent association of hypospadias with low birth weight, maternal hypertension, and preeclampsia suggests that placental insufficiency is a major risk factor. Maternal exposure to chemical pollutants or endocrine disruptors in high concentrations related to selected occupations or geographic areas may be additional risk factors for hypospadias, especially in genetically predisposed individuals. So far, however, no environmental chemical pollutants or endocrine disruptor with a general common impact on the risk for hypospadias in most societies has been demonstrated. SUMMARY: A major point that should be considered regarding the action of environmental toxicants in inducing hypospadias is the cumulative effects of multiple low-dose exposures. Furthermore, interactions between genetic and environmental factors may help to explain nonreplication in genetic studies of hypospadias.


Assuntos
Poluentes Ambientais/efeitos adversos , Interação Gene-Ambiente , Hipospadia/etiologia , Exposição Materna , Insuficiência Placentária/genética , Efeitos Tardios da Exposição Pré-Natal/genética , Adulto , Androgênios/deficiência , Gonadotropina Coriônica/metabolismo , Receptor alfa de Estrogênio/genética , Feminino , Predisposição Genética para Doença , Humanos , Hipospadia/epidemiologia , Hipospadia/genética , Recém-Nascido , Masculino , Exposição Materna/efeitos adversos , Insuficiência Placentária/etiologia , Polimorfismo de Nucleotídeo Único , Gravidez , Efeitos Tardios da Exposição Pré-Natal/etiologia , Fatores de Risco
12.
PLoS One ; 8(1): e54013, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23326560

RESUMO

OBJECTIVES: Recent studies have shown that telomere length was significantly reduced in placentas collected at delivery from pregnancies complicated by intrauterine growth restriction secondary to placental insufficiency. Placental telomere length measurement during ongoing pregnancies complicated by intrauterine growth restriction has never been reported. This was the main objective of our study. METHODS: In our center, late chorionic villus samplings were performed between 18 and 37 weeks of amenorrhea in 24 subjects with severe intrauterine growth restriction (cases) and in 28 subjects with other indications for prenatal diagnosis (controls). Placental insufficiency was assessed by histo-pathological examination. Relative measurement of telomere length was carried out prospectively by quantitative Fluorescent In Situ Hybridization using fluorescent Peptide Nucleic Acid probes on interphase nuclei obtained from long-term cultured villi and with an automated epifluorescent microscope. A quantitative Polymerase Chain Reaction technique was performed to confirm the quantitative Fluorescent In Situ Hybridization results. The number of copies of gene loci encoding the RNA template (hTERC) and the catalytic subunit (hTERT) of the enzyme complex telomerase were also estimated in these placentas by Fluorescent In Situ Hybridization. RESULTS: Mean fluorescence intensity of telomere probes estimated by quantitative Fluorescent In Situ Hybridization was significantly less for cases compared to controls (p<0.001). This result indicated that mean telomere length was significantly reduced in placentas during pregnancies complicated by intrauterine growth restriction. Reduced telomere length was confirmed by the quantitative Polymerase Chain Reaction technique. No copy number variation of the hTERC and hTERT loci was noticed for cases, or for controls. CONCLUSION: This study clearly demonstrates a reduction of placental telomere length in ongoing pregnancies (from 18 to 37 weeks of amenorrhea) complicated by severe intrauterine growth restriction secondary to placental insufficiency.


Assuntos
Retardo do Crescimento Fetal , Placenta/citologia , Insuficiência Placentária , Homeostase do Telômero/genética , Adulto , Amostra da Vilosidade Coriônica , Feminino , Retardo do Crescimento Fetal/fisiopatologia , Dosagem de Genes , Humanos , Hibridização in Situ Fluorescente , Placenta/fisiopatologia , Insuficiência Placentária/genética , Insuficiência Placentária/fisiopatologia , Gravidez , Complicações na Gravidez/fisiopatologia , RNA/genética , Telomerase/genética
13.
DNA Cell Biol ; 29(6): 295-301, 2010 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-20235876

RESUMO

This study evaluated quantification of fetal extracellular DNA in maternal plasma for differentiation between cases at risk of onset of placental-insufficiency-related complications and normal pregnancies. Using real-time polymerase chain reaction, fetal (sex-determining region Y [SRY] and hypermethylated RASSF1A sequence) and total (beta-globin [GLO] gene) extracellular DNA was examined in 70 normal pregnancies, 18 at risk of placental-insufficiency-related pregnancy complications, 24 preeclampsia with or without (w or w/o) intrauterine growth retardation (IUGR) (median 34.0 week), and 11 IUGR (median 28.5 week). IUGR was diagnosed when estimated fetal weight was below the 10th percentile for evaluated gestational age. Although increased levels of extracellular DNA were detected in pregnancies with preeclampsia w or w/o IUGR relative to controls (RASSF1A, p < 0.001; SRY, p = 0.009; GLO, p < 0.001), quantities of fetal extracellular DNA in IUGR were not statistically significant (RASSF1A, p = 0.21; SRY, p = 0.2). RASSF1A, SRY, and GLO achieved 93.1%, 93.6%, and 92.1% accuracy for differentiation between normal pregnancy and preeclampsia w or w/o IUGR. Lower sensitivity was observed for pregnancies with onset of IUGR (RASSF1A, 60.0%; SRY, 80.0%; GLO, 72.7%), but did not influence final accuracy (RASSF1A, 91.6%; SRY, 92.5%; GLO, 89.5%). Among 18 patients at risk, 8 pregnancies involving 3 female and 5 male fetuses developed preeclampsia (n = 4), IUGR (n = 3), and chronic placentopathy causing hypoxia (n = 1). Elevation of extracellular DNA was demonstrated in 3/5 (SRY), 1/8 (hypermethylated RASSF1A), and 4/8 (GLO) patients at the earliest 26 weeks and at the latest 2 weeks before the onset of symptoms. These data indicate that fetal and total extracellular DNA concentrations can be significantly elevated in plasma of patients who later developed placental-insufficiency-related pregnancy complications. However, this is strongly individualized, and not a rule for all cases, and probably depends on the actual occurrence of excessive placental trophoblast apoptosis.


Assuntos
Metilação de DNA , DNA/genética , Espaço Extracelular/genética , Insuficiência Placentária/diagnóstico , Proteína da Região Y Determinante do Sexo/genética , Proteínas Supressoras de Tumor/genética , Globinas beta/genética , Estudos de Casos e Controles , Feminino , Feto/citologia , Marcadores Genéticos/genética , Humanos , Masculino , Mães , Insuficiência Placentária/sangue , Insuficiência Placentária/genética , Gravidez , Risco , Proteína da Região Y Determinante do Sexo/sangue , Fatores de Tempo , Proteínas Supressoras de Tumor/sangue , Globinas beta/metabolismo
14.
Pediatr Res ; 67(6): 603-8, 2010 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-20220548

RESUMO

Low birth weight and catch-up growth predict increased adiposity in children and adults. This may be due in part to leptin resistance, as adults who were born small exhibit increased plasma leptin concentration relative to adiposity. Placental restriction (PR), a major cause of intrauterine growth restriction, reduces size at birth and increases feeding activity and adiposity by 6 wk in sheep. We hypothesized that PR would increase plasma leptin concentration and alter its relationship with feeding activity and adiposity in young lambs. Body size, plasma leptin, feeding activity, adiposity, leptin, and leptin receptor gene expression in adipose tissue were measured (12 control, 12 PR). PR reduced size at birth and increased adiposity. Plasma leptin concentration decreased with age, but to a lesser extent after PR and correlated positively with adiposity similarly in control and PR. PR increased plasma leptin concentration and perirenal adipose tissue leptin expression. Feeding activity correlated negatively with plasma leptin concentration in controls, but positively after PR. PR increases adipose tissue leptin expression and plasma leptin concentration, however, this increased abundance of peripheral leptin does not inhibit feeding activity (suckling event frequency), suggesting PR programs resistance to appetite and energy balance regulation by leptin, leading to early onset obesity.


Assuntos
Tecido Adiposo/metabolismo , Adiposidade , Comportamento Alimentar , Retardo do Crescimento Fetal/etiologia , Hiperfagia/etiologia , Leptina/sangue , Insuficiência Placentária/sangue , Fatores Etários , Animais , Animais Recém-Nascidos , Animais Lactentes , Peso ao Nascer , Glicemia/metabolismo , Modelos Animais de Doenças , Ácidos Graxos não Esterificados/sangue , Feminino , Retardo do Crescimento Fetal/sangue , Retardo do Crescimento Fetal/fisiopatologia , Hiperfagia/sangue , Hiperfagia/fisiopatologia , Insulina/sangue , Lactação , Leptina/genética , Masculino , Insuficiência Placentária/genética , Insuficiência Placentária/fisiopatologia , Gravidez , Receptores para Leptina/metabolismo , Ovinos , Regulação para Cima
15.
J Am Soc Nephrol ; 20(4): 777-86, 2009 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19297558

RESUMO

A number of studies have shown that placental insufficiency affects embryonic patterning of the kidney and leads to a decreased number of functioning nephrons in adulthood; however, there is circumstantial evidence that placental insufficiency may also affect renal medullary growth, which could account for cases of unexplained renal medullary dysplasia and for abnormalities in renal function among infants who had experienced intrauterine growth retardation. We observed that mice with late gestational placental insufficiency associated with genetic loss of Cited1 expression in the placenta had renal medullary dysplasia. This was not caused by lower urinary tract obstruction or by defects in branching of the ureteric bud during early nephrogenesis but was associated with decreased tissue oxygenation and increased apoptosis in the expanding renal medulla. Loss of placental Cited1 was required for Cited1 mutants to develop renal dysplasia, and this was not dependent on alterations in embryonic Cited1 expression. Taken together, these findings suggest that renal medullary dysplasia in Cited1 mutant mice is a direct consequence of decreased tissue oxygenation resulting from placental insufficiency.


Assuntos
Nefropatias/genética , Proteínas Nucleares/deficiência , Proteínas Nucleares/genética , Insuficiência Placentária/genética , Transativadores/deficiência , Fatores de Transcrição/deficiência , Fatores de Transcrição/genética , Animais , Proteínas Reguladoras de Apoptose , Feminino , Retardo do Crescimento Fetal/genética , Expressão Gênica , Humanos , Lactente , Rim/embriologia , Nefropatias/patologia , Medula Renal/embriologia , Medula Renal/patologia , Camundongos , Camundongos Endogâmicos C57BL , Placenta/fisiologia , Gravidez , Transativadores/genética , Transcrição Gênica
16.
Physiol Genomics ; 25(1): 16-28, 2006 Mar 13.
Artigo em Inglês | MEDLINE | ID: mdl-16380407

RESUMO

Intrauterine growth retardation (IUGR) increases the risk of neuroendocrine reprogramming. In the rat, IUGR leads to persistent changes in cerebral mRNA levels. This suggests lasting alterations in IUGR cerebral transcriptional regulation, which may result from changes in chromatin structure. Candidate nutritional triggers for these changes include altered cerebral zinc and one-carbon metabolite levels. We hypothesized that IUGR affects cerebral chromatin structure in neonatal and postnatal rat brains. Rats were rendered IUGR by bilateral uterine artery ligation; controls (Con) underwent sham surgery. At day of life 0 (d0), we measured cerebral DNA methylation, histone acetylation, expression of chromatin-affecting enzymes, and cerebral levels of one-carbon metabolites and zinc. At day of life 21 (d21), we measured cerebral DNA methylation and histone acetylation, as well as the caloric content of Con and IUGR rat breast milk. At d0, IUGR significantly decreased genome-wide and CpG island methylation, as well as increased histone 3 lysine 9 (H3/K9) and histone 3 lysine 14 (H3/K14) acetylation in the hippocampus and periventricular white matter, respectively. IUGR also decreased expression of the chromatin-affecting enzymes DNA methyltransferase 1 (DNMT1), methyl-CpG binding protein 2 (MeCP2), and histone deacetylase (HDAC)1 in association with increased cerebral levels of zinc. In d21 female IUGR rats, cerebral CpG DNA methylation remained lower, whereas H3/K9 and H3/K14 hyperacetylation persisted in hippocampus and white matter, respectively. In d21 male rats, IUGR decreased acetylation of H3/K9 and H3/K14 in these respective regions compared with controls. Despite these differences, caloric, fat, and protein content were similar in breast milk from Con and IUGR dams. We conclude that IUGR results in postnatal changes in cerebral chromatin structure and that these changes are sex specific.


Assuntos
Encéfalo/enzimologia , Cromatina/química , Epigênese Genética , Retardo do Crescimento Fetal/enzimologia , Insuficiência Placentária/enzimologia , Acetilação , Animais , Animais Recém-Nascidos , Encéfalo/ultraestrutura , Cromatina/genética , Cromatina/metabolismo , Ilhas de CpG , DNA/genética , DNA/metabolismo , DNA (Citosina-5-)-Metiltransferase 1 , DNA (Citosina-5-)-Metiltransferases/genética , DNA (Citosina-5-)-Metiltransferases/metabolismo , Metilação de DNA , Feminino , Retardo do Crescimento Fetal/genética , Histona Desacetilases/genética , Histona Desacetilases/metabolismo , Histonas/metabolismo , Imuno-Histoquímica , Masculino , Proteína 2 de Ligação a Metil-CpG/genética , Proteína 2 de Ligação a Metil-CpG/metabolismo , Estrutura Molecular , Neurônios/enzimologia , Neurônios/ultraestrutura , Insuficiência Placentária/genética , Gravidez , RNA Mensageiro/metabolismo , Ratos , Fatores Sexuais , Zinco/metabolismo
17.
Am J Physiol Regul Integr Comp Physiol ; 289(5): R1348-53, 2005 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-16002560

RESUMO

Uteroplacental insufficiency (UPI) induces persistent changes in hepatic gene expression secondary to altered chromatin dynamics in the intrauterine growth- restricted (IUGR) rat liver. The glucocorticoid receptor (GR) is a transcription factor that when activated can induce changes in chromatin structure. To begin the process of identifying pathways by which IUGR affects chromatin structure, we hypothesized that UPI in the rat induces a significant increase in endogenous glucocorticoids (corticosterone) and increases GR expression and activation. To prove our hypothesis, we induced IUGR through bilateral uterine artery ligation of the pregnant rat. At day 1, UPI significantly increased corticosterone levels and was associated with increased total GR mRNA and protein levels in the liver, as well as increased hepatic phosphorylation of GR serine 211. Moreover, cyclin-dependent kinase 2 (CDK2) cyclinA/CDK2 protein levels, which selectively phosphorylate GR serine 211, were also significantly increased. To assess activity of the GR, we measured protein levels of the transcription factor p53 whose levels are downregulated, at least in part, by active GR. In this study, UPI decreased p53 protein and its downstream target Bax mRNA levels. We conclude that UPI in rats affects GR expression and activity in the liver. We speculate that these alterations early in life may contribute to the changes in chromatin structure and gene expression previously described in the IUGR liver.


Assuntos
Retardo do Crescimento Fetal/metabolismo , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Insuficiência Placentária/metabolismo , Receptores de Glucocorticoides/metabolismo , Animais , Cromatina/genética , Cromatina/metabolismo , Corticosterona/sangue , Ciclina A/metabolismo , Feminino , Retardo do Crescimento Fetal/etiologia , Retardo do Crescimento Fetal/genética , Fígado/metabolismo , Fosforilação , Insuficiência Placentária/genética , Gravidez , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Ratos , Ratos Sprague-Dawley , Serina/metabolismo , Proteína Supressora de Tumor p53/genética , Proteína Supressora de Tumor p53/metabolismo
18.
Early Hum Dev ; 79(2): 93-105, 2004 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-15324990

RESUMO

Human and animal studies demonstrate that uteroplacental insufficiency and subsequent intrauterine growth retardation (IUGR) decrease intestinal growth and lead to both an increased incidence of feeding intolerance and necrotizing enterocolitis. Our objective was to determine the effects of uteroplacental insufficiency upon small intestine growth, histology, gene expression of the apoptosis related proteins Bcl-2, Bax and p53, and caspase-3 activity. For this purpose, we induced uteroplacental insufficiency through bilateral uterine artery ligation on day 19 of gestation in fully anesthetized pregnant Sprague-Dawley rats and harvested pups at term 2 days latter. Pups from sham surgeries served as controls. Uteroplacental insufficiency reduced cell count per crypt and decreased small intestinal weight. In association with these changes, IUGR intestinal Bcl-2 mRNA levels were decreased significantly, and Bax and p53 mRNA were significantly increased in distal ileum. Immunohistochemistry for Bcl-2, Bax, and p53 revealed similar findings. In association with the decreased Bcl-2 and the increased Bax gene expression, increased caspase-3 activity characterized the IUGR distal ileum. We conclude that uteroplacental insufficiency affects intestinal growth and morphology in association with altered gene expression of apoptosis related proteins. We speculate that the morphological change and associated altered apoptotic homeostasis contribute to the increased morbidity of infants affected by uteroplacental insufficiency.


Assuntos
Apoptose , Retardo do Crescimento Fetal/metabolismo , Íleo/anormalidades , Jejuno/anormalidades , Placenta/patologia , Insuficiência Placentária/patologia , Animais , Apoptose/genética , Modelos Animais de Doenças , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Idade Gestacional , Homeostase/genética , Íleo/metabolismo , Técnicas Imunoenzimáticas , Jejuno/metabolismo , Placenta/irrigação sanguínea , Insuficiência Placentária/genética , Insuficiência Placentária/metabolismo , Gravidez , Proteínas Proto-Oncogênicas c-bcl-2/genética , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , RNA Mensageiro/metabolismo , Ratos , Ratos Sprague-Dawley , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Proteína Supressora de Tumor p53/genética , Proteína Supressora de Tumor p53/metabolismo , Proteína X Associada a bcl-2
19.
Physiol Genomics ; 18(1): 43-50, 2004 Jun 17.
Artigo em Inglês | MEDLINE | ID: mdl-15084713

RESUMO

Uteroplacental insufficiency leads to intrauterine growth retardation (IUGR) and increases the risk of insulin resistance and hypertriglyceridemia in both humans and rats. Postnatal changes in hepatic gene expression characterize the postnatal IUGR rat, despite the transient nature of the initial in utero insult. Phenomena such as DNA methylation and histone acetylation can induce a relatively static reprogramming of gene transcription by altering chromatin infrastructure. We therefore hypothesized that uteroplacental insufficiency persistently affects DNA methylation and histone acetylation in the IUGR rat liver. IUGR rat pups were created by inducing uteroplacental insufficiency through bilateral uterine artery ligation of the pregnant dam on day 19 of gestation. The SssI methyltransferase assay and two-dimensional thin-layer chromatography demonstrated genome-wide DNA hypomethylation in postnatal IUGR liver. To investigate a possible mechanism for this hypomethylation, levels of hepatic metabolites and enzyme mRNAs involved in one-carbon metabolism were measured using HPLC with coulometric electrochemical detection and real-time RT-PCR, respectively. Uteroplacental insufficiency increased IUGR levels of S-adenosylhomocysteine, homocysteine, and methionine in association with decreased mRNA levels of methionine adenosyltransferase and cystathionine-beta-synthase. Western blotting further demonstrated that increased quantities of acetylated histone H3 also characterized the IUGR liver. Increased hepatic levels of S-adenosylhomocysteine can promote DNA hypomethylation, which is often associated with histone hyperacetylation. We speculate that the altered intrauterine milieu associated with uteroplacental insufficiency affects hepatic one-carbon metabolism and subsequent DNA methylation, which thereby alters chromatin dynamics and leads to persistent changes in hepatic gene expression.


Assuntos
Carbono/metabolismo , Metilação de DNA , Retardo do Crescimento Fetal/metabolismo , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Histonas/metabolismo , Circulação Placentária , Insuficiência Placentária/metabolismo , Processamento de Proteína Pós-Traducional , Acetilação , Animais , Cromatina/genética , Cistationina beta-Sintase/biossíntese , Cistationina beta-Sintase/genética , Modelos Animais de Doenças , Suscetibilidade a Doenças , Indução Enzimática , Feminino , Retardo do Crescimento Fetal/etiologia , Retardo do Crescimento Fetal/genética , Idade Gestacional , Fígado/metabolismo , Metionina/metabolismo , Metionina Adenosiltransferase/biossíntese , Metionina Adenosiltransferase/genética , Insuficiência Placentária/genética , Gravidez , Ratos , S-Adenosil-Homocisteína/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA