Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Mol Hum Reprod ; 25(9): 572-585, 2019 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-31418778

RESUMEN

Fetal growth restriction (FGR) is caused by poor placental development and function early in gestation. It is well known that placentas from women with FGR exhibit reduced cell growth, elevated levels of apoptosis and perturbed expression of the growth factors, cytokines and the homeobox gene family of transcription factors. Previous studies have reported that insulin-like growth factor-2 (IGF2) interacts with its receptor-2 (IGF2R) to regulate villous trophoblast survival and apoptosis. In this study, we hypothesized that human placental IGF2R-mediated homeobox gene expression is altered in FGR and contributes to abnormal trophoblast function. This study was designed to determine the association between IGF2R, homeobox gene expression and cell survival in pregnancies affected by FGR. Third trimester placentas were collected from FGR-affected pregnancies (n = 29) and gestation matched with control pregnancies (n = 30). Functional analyses were then performed in vitro using term placental explants (n = 4) and BeWo trophoblast cells. mRNA expression was determined by real-time PCR, while protein expression was examined by immunoblotting and immunohistochemistry. siRNA transfection was used to silence IGF2R expression in placental explants and the BeWo cell-line. cDNA arrays were used to screen for downstream targets of IGF2R, specifically homeobox gene transcription factors and apoptosis-related genes. Functional effects of silencing IGF2R were then verified by ß-hCG ELISA, caspase activity assays and a real-time electrical cell-impedance assay for differentiation, apoptosis and cell growth potential, respectively. IGF2R expression was significantly decreased in placentas from pregnancies complicated by idiopathic FGR (P < 0.05 versus control). siRNA-mediated IGF2R knockdown in term placental explants and the trophoblast cell line BeWo resulted in altered expression of homeobox gene transcription factors, including increased expression of distal-less homeobox gene 5 (DLX5), and decreased expression of H2.0-Like Homeobox 1 (HLX) (P < 0.05 versus control). Knockdown of IGF2R transcription increased the expression and activity of caspase-6 and caspase-8 in placental explants, decreased BeWo proliferation and increased BeWo differentiation (all P < 0.05 compared to respective controls). This is the first study linking IGF2R placental expression with changes in the expression of homeobox genes that control cellular signalling pathways responsible for increased trophoblast cell apoptosis, which is a characteristic feature of FGR.


Asunto(s)
Apoptosis/genética , Retardo del Crecimiento Fetal/genética , Genes Homeobox , Proteínas de Homeodominio/genética , Placenta/metabolismo , Receptor IGF Tipo 2/fisiología , Adulto , Estudios de Casos y Controles , Línea Celular , Femenino , Retardo del Crecimiento Fetal/patología , Expresión Génica , Humanos , Recién Nacido , Placenta/patología , Placentación/genética , Embarazo
2.
Biochimie ; 161: 80-87, 2019 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-30605696

RESUMEN

Fetal growth restriction (FGR) is a complex disorder of human pregnancy that leads to poor health outcomes in offspring. These range from immediate risks such as perinatal morbidity and stillbirths, to long-term complications including severe neurodevelopmental problems. Despite its relatively high global prevalence, the aetiology of FGR and its complications is not currently well understood. We now know that serotonin (5-HT) is synthesised in the placenta and is crucial for early fetal forebrain development in mice. However, the contribution of a disrupted placental 5-HT synthetic pathway to the pathophysiology of placental insufficiency in FGR and its significant fetal neurodevelopmental complications are unclear.


Asunto(s)
Retardo del Crecimiento Fetal/fisiopatología , Placenta/fisiopatología , Insuficiencia Placentaria , Receptores de Serotonina/metabolismo , Serotonina/metabolismo , Femenino , Retardo del Crecimiento Fetal/metabolismo , Humanos , Placenta/metabolismo , Embarazo
3.
Mol Hum Reprod ; 24(2): 94-109, 2018 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-29272530

RESUMEN

STUDY QUESTION: What is the association between placental formyl peptide receptor 2 (FPR2) and trophoblast and endothelial functions in pregnancies affected by foetal growth restriction (FGR)? SUMMARY ANSWER: Reduced FPR2 placental expression in idiopathic FGR results in significantly altered trophoblast differentiation and endothelial function in vitro. WHAT IS KNOWN ALREADY: FGR is associated with placental insufficiency, where defective trophoblast and endothelial functions contribute to reduced feto-placental growth. STUDY DESIGN, SIZE, DURATION: The expression of FPR2 in placental tissues from human pregnancies complicated with FGR was compared to that in gestation-matched uncomplicated control pregnancies (n = 25 from each group). Fpr2 expression was also determined in placental tissues obtained from a murine model of FGR (n = 4). The functional role of FPR2 in primary trophoblasts and endothelial cells in vitro was assessed in diverse assays in a time-dependent manner. PARTICIPANTS/MATERIALS, SETTING, METHODS: Placentae from third-trimester pregnancies complicated by idiopathic FGR (n = 25) and those from gestation-matched pregnancies with appropriately grown infants as controls (n = 25) were collected at gestation 27-40 weeks. Placental tissues were also collected from a spontaneous CBA/CaH × DBA/2 J murine model of FGR. Placental FPR2/Fpr2 mRNA expression was determined by real-time PCR, while protein expression was examined by immunoblotting and immunohistochemistry. siRNA transfection was used to silence FPR2 expression in primary trophoblasts and in human umbilical vein endothelial cells (HUVEC), and the quantitation of cytokines, chemokines and apoptosis was performed following a cDNA array analyses. Functional effects of trophoblast differentiation were measured using HCGB/ß-hCG and syncytin-2 expression as well as markers of apoptosis, tumour protein 53 (TP53), caspase 8, B cell lymphoma 2 (BCL2) and BCL associated X (BAX). Endothelial function was assessed by proliferation, network formation and permeability assays. MAIN RESULTS AND THE ROLE OF CHANCE: Placental FPR2/Fpr2 expression was significantly decreased in FGR placentae (n = 25, P < 0.05) as well as in murine FGR placentae compared to controls (n = 4, P < 0.05). FPR2 siRNA (siFPR2) in term trophoblasts significantly increased differentiation markers, HCGB and syncytin-2; cytokines, interleukin (IL)-6, CXCL8; and apoptotic markers, TP53, caspase 8 and BAX, but significantly reduced the expression of the chemokines CXCL12 and its receptors CXCR4 and CXCR7; CXCL16 and its receptor, CXCR6; and cytokine, IL-10, compared with control siRNA (siCONT). Treatment of HUVECs with siFPR2 significantly reduced proliferation and endothelial tube formation, but significantly increased permeability of HUVECs. LARGE SCALE DATA: N/A. LIMITATIONS, REASONS FOR CAUTION: Reduced expression of placental FPR2/Fpr2 was observed in the third trimester at delivery after development of FGR, suggesting that FPR2 is associated with FGR pregnancies. However, there is a possibility that the decreased placental FPR2 observed in FGR may be a consequence rather than a cause of FGR, although our in vitro functional analyses using primary trophoblasts and endothelial cells suggest that FPR2 may have a direct or indirect regulatory role on trophoblast differentiation and endothelial function in FGR. WIDER IMPLICATIONS OF THE FINDINGS: This is the first study linking placental FPR2 expression with changes in the trophoblast and endothelial functions that may explain the placental insufficiency observed in FGR. STUDY FUNDING/COMPETING INTERESTS: P.M. and P.R.E. received funding from the Australian Institute of Musculoskeletal Science, Western Health, St. Albans, Victoria 3021, Australia. M.L. is supported by a Career Development Fellowship from the National Health and Medical Research Council (NHMRC; Grant no. 1047025). Monash Health is supported by the Victorian Government's Operational Infrastructure Support Programme. The authors declare that there is no conflict of interest in publishing this work.


Asunto(s)
Retardo del Crecimiento Fetal/metabolismo , Placenta/metabolismo , Receptores de Formil Péptido/metabolismo , Receptores de Lipoxina/metabolismo , Apoptosis/genética , Apoptosis/fisiología , Diferenciación Celular/genética , Diferenciación Celular/fisiología , Células Endoteliales/citología , Células Endoteliales/metabolismo , Femenino , Células Endoteliales de la Vena Umbilical Humana/citología , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Humanos , Embarazo , Primer Trimestre del Embarazo , Tercer Trimestre del Embarazo , Receptores de Formil Péptido/genética , Receptores de Lipoxina/genética , Transducción de Señal/genética , Transducción de Señal/fisiología , Trofoblastos/citología , Trofoblastos/metabolismo
4.
Twin Res Hum Genet ; 21(1): 42-50, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-29212571

RESUMEN

A discordant twin gestation, in which one fetus is significantly growth restricted, compared to the other normal twin, is a unique model that can be used to elucidate the mechanism(s) by which the intrauterine environment affects fetal growth. In many model systems, placental transcription factor genes regulate fetal growth. Transcription factors regulate growth through their activation or repression of downstream target genes that mediate important cell functions. The objective of this study was to determine the expression of the placental HLX homeobox gene transcription factor and its downstream target genes in dizygotic twins with growth discordance. In this cross-sectional study, HLX and its downstream target genes' retinoblastoma 1 (RB1) and cyclin kinase D (CDKN1C) expression levels were determined in placentae obtained from dichorionic diamniotic twin pregnancies (n = 23) where one of the twins was growth restricted. Fetal growth restriction (FGR) was defined as small for gestational age with abnormal umbilical artery Doppler indices when compared with the normal control co-twin. Homeobox gene HLX expression was significantly decreased at both the mRNA and protein levels in FGR twin placentae compared with the normal control co-twin placentae (p < .05). Downstream target genes CDKN1C and RB1 were also significantly decreased and increased, respectively, at both the mRNA and protein levels in FGR twin placentae compared with normal control co-twin placentae (p < .05). Together, these observations suggest an important association between HLX transcription factor expression and abnormal human placental development in discordant twin pregnancies.


Asunto(s)
Retardo del Crecimiento Fetal/genética , Proteínas de Homeodominio/genética , Placenta/fisiología , Embarazo Gemelar/genética , Factores de Transcripción/genética , Peso al Nacer , Inhibidor p57 de las Quinasas Dependientes de la Ciclina/genética , Femenino , Regulación del Desarrollo de la Expresión Génica , Proteínas de Homeodominio/metabolismo , Humanos , Inmunohistoquímica , Embarazo , Proteínas de Unión a Retinoblastoma/genética , Factores de Transcripción/metabolismo , Ubiquitina-Proteína Ligasas/genética
5.
Arterioscler Thromb Vasc Biol ; 37(6): 1168-1179, 2017 06.
Artículo en Inglés | MEDLINE | ID: mdl-28408374

RESUMEN

OBJECTIVE: Biglycan (BGN) has reduced expression in placentae from pregnancies complicated by fetal growth restriction (FGR). We used first trimester placental samples from pregnancies with later small for gestational age (SGA) infants as a surrogate for FGR. The functional consequences of reduced BGN and the downstream targets of BGN were determined. Furthermore, the expression of targets was validated in primary placental endothelial cells isolated from FGR or control pregnancies. APPROACH AND RESULTS: BGN expression was determined using real-time polymerase chain reaction in placental tissues collected during chorionic villous sampling performed at 10 to 12 weeks' gestation from pregnancies that had known clinical outcomes, including SGA. Short-interference RNA reduced BGN expression in telomerase-immortalized microvascular endothelial cells, and the effect on proliferation, angiogenesis, and thrombin generation was determined. An angiogenesis array identified downstream targets of BGN, and their expression in control and FGR primary placental endothelial cells was validated using real-time polymerase chain reaction. Reduced BGN expression was observed in SGA placental tissues. BGN reduction decreased network formation of telomerase-immortalized microvascular endothelial cells but did not affect thrombin generation or cellular proliferation. The array identified target genes, which were further validated: angiopoetin 4 (ANGPT4), platelet-derived growth factor receptor α (PDGFRA), tumor necrosis factor superfamily member 15 (TNFSF15), angiogenin (ANG), serpin family C member 1 (SERPIN1), angiopoietin 2 (ANGPT2), and CXC motif chemokine 12 (CXCL12) in telomerase-immortalized microvascular endothelial cells and primary placental endothelial cells obtained from control and FGR pregnancies. CONCLUSIONS: This study reports a temporal relationship between altered placental BGN expression and subsequent development of SGA. Reduction of BGN in vascular endothelial cells leads to disrupted network formation and alterations in the expression of genes involved in angiogenesis. Therefore, differential expression of these may contribute to aberrant angiogenesis in SGA pregnancies.


Asunto(s)
Biglicano/metabolismo , Vellosidades Coriónicas/irrigación sanguínea , Vellosidades Coriónicas/metabolismo , Células Endoteliales/metabolismo , Retardo del Crecimiento Fetal/metabolismo , Microvasos/metabolismo , Neovascularización Fisiológica , Primer Trimestre del Embarazo/metabolismo , Telomerasa/metabolismo , Animales , Biglicano/genética , Estudios de Casos y Controles , Línea Celular , Muestra de la Vellosidad Coriónica , Femenino , Retardo del Crecimiento Fetal/genética , Retardo del Crecimiento Fetal/fisiopatología , Perfilación de la Expresión Génica , Regulación del Desarrollo de la Expresión Génica , Humanos , Recién Nacido , Recién Nacido Pequeño para la Edad Gestacional , Masculino , Ratones Endogámicos C57BL , Neovascularización Fisiológica/genética , Embarazo , Primer Trimestre del Embarazo/genética , Interferencia de ARN , Transducción de Señal , Telomerasa/genética , Trombina/metabolismo , Factores de Tiempo , Técnicas de Cultivo de Tejidos , Transfección
6.
Am J Pathol ; 186(12): 3217-3224, 2016 12.
Artículo en Inglés | MEDLINE | ID: mdl-27750048

RESUMEN

Preeclampsia (PE), a serious hypertensive disorder of pregnancy, remains a leading cause of perinatal morbidity and mortality worldwide. Perturbed trophoblast function and impaired placental development early in pregnancy are key features. Low-dose acetylsalicylic acid (LDA) administered before 16 weeks' gestation significantly reduces the risk for PE. However, the exact mechanisms of action of LDA, particularly on trophoblast function, are unclear. We hypothesized that LDA influences placental trophoblast function and reverses PE-associated abnormalities. This study aimed to determine the effects of serum from normotensive women and from those with PE with or without LDA treatment on a model of placental syncytium. On cytokine profiling, LDA increased placental growth factor production and selectively restored PE serum-induced alterations in levels of cytokines [activated leukocyte cell adhesion molecule, CXCL-16, and ErbB3] to those in normotensive serum-treated cells. PE serum-induced increases in the apoptotic markers P53 mRNA expression, IKBKE mRNA expression, caspase 3 activity, and decreased BIRC8 mRNA expression, were attenuated by LDA treatment. LDA treatment also reduced abnormal differentiation caused by PE serum administration. Possible mechanisms by which LDA influences PE-affected trophoblast cells in vitro are by modulating cytokine secretion, reducing apoptosis to levels seen in normotensive serum-treated cells, and preventing the premature trophoblast differentiation commonly observed in PE.


Asunto(s)
Aspirina/administración & dosificación , Citocinas/metabolismo , Preeclampsia/tratamiento farmacológico , Trofoblastos/metabolismo , Apoptosis/efectos de los fármacos , Línea Celular , Femenino , Células Gigantes/metabolismo , Humanos , Placenta/metabolismo , Factor de Crecimiento Placentario/metabolismo , Preeclampsia/metabolismo , Embarazo
7.
Reproduction ; 152(5): 457-65, 2016 11.
Artículo en Inglés | MEDLINE | ID: mdl-27539603

RESUMEN

Aberrant placental angiogenesis is associated with fetal growth restriction (FGR). In mice, targeted disruption of the homeobox gene, transforming growth ß-induced factor (Tgif-1), which is also a transcription factor, causes defective placental vascularisation. Nevertheless, the role of TGIF-1 in human placental angiogenesis is unclear. We have previously reported increased TGIF-1 expression in human FGR placentae and demonstrated localisation of TGIF-1 protein in placental endothelial cells (ECs). However, its functional role remains to be investigated. In this study, we aimed to specifically compare TGIF-1 mRNA expression in placental ECs isolated from human FGR-affected pregnancies with gestation-matched control pregnancies in two independent cohorts from Australia and Canada and to identify the functional role of TGIF-1 in placental angiogenesis using the human umbilical vein endothelial cell-derived cell line, SGHEC-7, and primary human umbilical vein ECs. Real-time PCR revealed that TGIF-1 mRNA expression was significantly increased in ECs isolated from FGR-affected placentae compared with that of controls. The functional roles of TGIF-1 were determined in ECs after TGIF-1 siRNA transfection. TGIF-1 inactivation in ECs significantly reduced TGIF-1 at both the mRNA and protein levels, as well as the proliferative and invasive potential, but significantly increased the angiogenic potential. Using angiogenesis PCR screening arrays, we identified ITGAV, NRP-1, ANPGT-1 and ANPGT-2 as novel downstream targets of TGIF-1, after TGIF-1 inactivation in ECs. Collectively, these results show that TGIF-1 regulates EC function and the expression of angiogenic molecules; and when abnormally expressed, may contribute to the aberrant placental angiogenesis observed in FGR.


Asunto(s)
Células Endoteliales/patología , Retardo del Crecimiento Fetal/diagnóstico , Retardo del Crecimiento Fetal/metabolismo , Proteínas de Homeodominio/metabolismo , Placenta/patología , Proteínas Represoras/metabolismo , Trofoblastos/patología , Adulto , Proliferación Celular , Células Cultivadas , Células Endoteliales/metabolismo , Femenino , Humanos , Lactante , Masculino , Placenta/metabolismo , Embarazo , Trofoblastos/metabolismo
8.
Angiogenesis ; 18(2): 115-23, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25433512

RESUMEN

Placental angiogenesis is critical to the success of human pregnancy. Angiogenesis is defined as the formation of new blood vessels from existing vasculature. Angiogenesis is necessary for the establishment of adequate placental perfusion, which is important for providing the optimum in utero environment to support fetal development. Defective placental angiogenesis is associated with several pregnancy complications, the most clinically important of which is preeclampsia; the multisystem disorder is characterized by maternal hypertension, proteinuria, and endothelial dysfunction. Here, we review our current understanding of several key angiogenic factors that are associated with placental angiogenesis. We also discuss their importance with respect to preeclampsia, where aberrant expression and release of these factors into the maternal circulation is thought to contribute to the pathogenesis and pathophysiology of preeclampsia.


Asunto(s)
Proteínas Angiogénicas/fisiología , Placenta/fisiología , Preeclampsia/etiología , Femenino , Humanos , Embarazo
9.
Reprod Sci ; 21(11): 1382-94, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-24692208

RESUMEN

Human chorionic mesenchymal stem/stromal cells (CMSCs) derived from the placenta are similar to adult tissue-derived MSCs. The aim of this study was to investigate the role of these cells in normal placental development. Transcription factors, particularly members of the homeobox gene family, play crucial roles in maintaining stem cell proliferation and lineage specification in embryonic tissues. In adult tissues and organs, stem cells proliferate at low levels in their niche until they receive cues from the microenvironment to differentiate. The homeobox genes that are expressed in the CMSC niche in placental tissues have not been identified. We used the novel strategy of laser capture microdissection to isolate the stromal component of first trimester villi and excluded the cytotrophoblast and syncytiotrophoblast layers that comprise the outer layer of the chorionic villi. Microarray analysis was then used to screen for homeobox genes in the microdissected tissue. Candidate homeobox genes were selected for further RNA analysis. Immunohistochemistry of candidate genes in first trimester placental villous stromal tissue revealed homeobox genes Meis1, myeloid ectropic viral integration site 1 homolog 2 (MEIS2), H2.0-like Drosophila (HLX), transforming growth factor ß-induced factor (TGIF), and distal-less homeobox 5 (DLX5) were expressed in the vascular niche where CMSCs have been shown to reside. Expression of MEIS2, HLX, TGIF, and DLX5 was also detected in scattered stromal cells. Real-time polymerase chain reaction and immunocytochemistry verified expression of MEIS2, HLX, TGIF, and DLX5 homeobox genes in first trimester and term CMSCs. These data suggest a combination of regulatory homeobox genes is expressed in CMSCs from early placental development to term, which may be required for stem cell proliferation and differentiation.


Asunto(s)
Corion/metabolismo , Proteínas de Homeodominio/metabolismo , Células Madre Mesenquimatosas/metabolismo , Diferenciación Celular , Proliferación Celular , Células Cultivadas , Corion/citología , Femenino , Perfilación de la Expresión Génica , Regulación del Desarrollo de la Expresión Génica , Genes Homeobox , Proteínas de Homeodominio/genética , Humanos , Inmunohistoquímica , Captura por Microdisección con Láser , Embarazo , Primer Trimestre del Embarazo , Segundo Trimestre del Embarazo , Reacción en Cadena en Tiempo Real de la Polimerasa , Proteínas Represoras/genética , Proteínas Represoras/metabolismo , Nicho de Células Madre , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
10.
Obstet Gynecol ; 115(1): 5-13, 2010 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-20027027

RESUMEN

OBJECTIVE: To estimate the association between five commonly inherited thrombophilia polymorphisms and adverse pregnancy outcomes in women who had no prior history of adverse pregnancy outcomes or personal or family history of venous thromboembolism. METHODS: Healthy nulliparous women (n=2,034) were recruited to this prospective cohort study before 22 weeks of gestation. Genotyping for factor V Leiden, prothrombin gene mutation, methylenetetrahydrofolate reductase enzyme (MTHFR) C677T, MTHFR A1298C, and thrombomodulin polymorphism was performed. Clinicians caring for women were blinded to the results of thrombophilia tests. The primary composite outcome was the development of severe preeclampsia, fetal growth restriction, placental abruption, stillbirth, or neonatal death. RESULTS: Complete molecular results and pregnancy outcome data were available in 1,707 women. These complications were experienced by 136 women (8.0%). Multivariable logistic regression demonstrated two statistically significant findings. Women who carried the prothrombin gene mutation had an odds ratio of 3.58 (95% confidence interval [CI] 1.20-10.61, P=.02) for the development of the composite primary outcome. Homozygous carriers of the MTHFR 1298 polymorphism had an odds ratio of 0.26 (95% CI 0.08-0.86, P=.03). None of the other polymorphisms studied showed a significant association with the development of the primary outcome in this cohort of women. CONCLUSION: Prothrombin gene mutation confers an increased risk for the development of adverse pregnancy outcomes in otherwise asymptomatic, nulliparous women, whereas homozygosity for MTHFR 1298 may protect against these complications. The majority of asymptomatic women who carry an inherited thrombophilia polymorphism have a successful pregnancy outcome. LEVEL OF EVIDENCE: II.


Asunto(s)
Factor V/genética , Metilenotetrahidrofolato Reductasa (NADPH2)/genética , Mutación Puntual/genética , Polimorfismo Genético , Resultado del Embarazo/genética , Protrombina/genética , Trombomodulina/genética , Trombofilia/genética , Desprendimiento Prematuro de la Placenta/epidemiología , Adulto , Femenino , Retardo del Crecimiento Fetal/epidemiología , Homocigoto , Humanos , Modelos Logísticos , Mutación , Preeclampsia/epidemiología , Embarazo , Resultado del Embarazo/epidemiología , Estudios Prospectivos , Medición de Riesgo , Mortinato/epidemiología , Trombofilia/epidemiología
11.
Aust N Z J Obstet Gynaecol ; 48(6): 536-41, 2008 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19133039

RESUMEN

AIM: Inherited thrombophilic polymorphisms have been linked to pregnancy-related thromboembolism and other adverse pregnancy outcomes. As there are limited data on the prevalence of these polymorphisms in Australian populations, we aimed to assess this in an antenatal population. METHODS: Healthy nulliparous women (n = 2031) were recruited to this study. The women had no past or family history of venous thromboembolism. Women were excluded if they or a family member was known to be a carrier of any thrombophilic marker. Genotyping from venous blood for the factor V Leiden, prothrombin 20210A, MTHFR 677 and 1298 and thrombomodulin C1418T polymorphisms was undertaken. RESULTS: Key findings were that 107 of 2019 (5.30, 95% confidence interval 4.36-6.37%) women tested were heterozygous carriers of factor V Leiden and one was homozygous (0.05, 0-0.27%); 2.43% of women were heterozygous carriers of the prothrombin gene mutation (1.80-3.20%) while no women were homozygous for this mutation; 11.62% (10.22-13.02%) and 9.98% (8.67-11.29%) were homozygous for the MTHFR 677 and 1298 polymorphisms, respectively, and 3.43% (2.63-4.22%) of women were homozygous for the thrombomodulin polymorphism. CONCLUSIONS: The prevalence of these polymorphisms is consistent with previously published data in Caucasian populations. These data will provide the basis for further assessment of the relationship between poor pregnancy outcome and these inherited thrombophilic polymorphisms in an asymptomatic antenatal population.


Asunto(s)
Trastornos de la Coagulación Sanguínea Heredados/epidemiología , Factor V/genética , Metilenotetrahidrofolato Reductasa (NADPH2)/genética , Polimorfismo Genético , Protrombina/genética , Trombomodulina/genética , Trombofilia/epidemiología , Australia/epidemiología , Trastornos de la Coagulación Sanguínea Heredados/etnología , Femenino , Frecuencia de los Genes , Predisposición Genética a la Enfermedad , Heterocigoto , Homocigoto , Humanos , Mutación , Embarazo , Resultado del Embarazo/genética , Prevalencia , Trombofilia/etnología , Trombofilia/genética
12.
Hum Mol Genet ; 16(12): 1504-14, 2007 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-17468176

RESUMEN

The mitochondria are the major cellular site of energy production and respiration. Recent research has focused on investigating the role of mitochondria in disease development and it has become increasingly evident that mitochondrial dysfunction contributes to a variety of human diseases. Mitochondrial DNA (mtDNA) quantity is very important for maintaining mitochondrial function and meeting the energy needs of the body. We have measured mitochondrial content in 1259 Mexican American individuals (from 42 extended families) and have shown that mtDNA quantity (a surrogate measure of mitochondrial integrity) has a large genetic component. We performed a genome scan and a genome-wide quantitative transcriptomic scan to identify QTLs influencing mitochondrial content. A variance components linkage-based genome scan utilizing 439 STR markers was used to localize a QTL for mitochondrial content on chromosome 10q (LOD = 3.83). Significant linkage to the mitochondrial genome was also detected for mitochondrial transmission (LOD = 3.39). For replication, we measured mitochondrial content in an independent Caucasian population (1088 individuals) finding evidence for linkage in these same regions. As part of the San Antonio Family Heart Study, we obtained genome-wide quantitative transcriptional profiles from 1240 individuals. Using lymphocyte samples, we quantitated 20 413 transcripts and examined correlations between the expression levels of these transcripts and mitochondrial content using the variance components method. Using regression analysis allowing for residual genetic components, we identified 829 transcripts (including many novel genes) influencing mitochondrial content that vary in their general biological actions, from cell signaling to cell trafficking and ion binding.


Asunto(s)
ADN Mitocondrial/metabolismo , Predisposición Genética a la Enfermedad , Mitocondrias/genética , Femenino , Ligamiento Genético , Marcadores Genéticos , Variación Genética , Humanos , Masculino , Americanos Mexicanos , Sitios de Carácter Cuantitativo , Análisis de Regresión , Transcripción Genética
13.
Hum Biol ; 78(4): 403-12, 2006 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-17278618

RESUMEN

Differences in the prevalence of thrombophilias in different ethnic populations have been demonstrated. Because the Australian population includes many different ethnic groups, we sought to assess the effect of ethnicity in our Australian prenatal population on the prevalence of thrombophilic polymorphisms. Asymptomatic, nulliparous women (n = 1,129) recruited for a large prospective study were included in this analysis. These women had no personal or family history of venous thromboembolism and were not known to be carrying an inherited or acquired thrombophilia. Ethnicity was determined at recruitment, and women were categorized as being of Northern European, Southern European, Middle Eastern, Asian, or Other ethnicity. These women underwent genotyping for the following polymorphisms: factor V Leiden G1691A, prothrombin gene A20210G mutation, methylenetetrahydrofolate reductase (MTHFR) C677T and A1298C, and thrombomodulin C1418T. The factor V Leiden allele was seen significantly more frequently in patients of Middle Eastern background compared to those of Northern European and Asian ethnicity (p < 0.05). The prothrombin gene mutation was seen significantly more frequently in patients of Southern European ethnicity compared to those of Northern European or Asian ethnicity (p < 0.05). The MTHFR C677T allele (mutant) was significantly less common in those of Asian ethnicity compared to patients of Northern European and Southern European ethnicity (p < 0.0005). There were no significant differences seen with the MTHFR A1298C polymorphism. The mutant thrombomodulin allele was seen significantly more frequently in Asian women compared to Northern European, Southern European, or Middle Eastern women (p < 0.005). There are important ethnic differences in the prevalence of thrombophilic polymorphisms in the Australian prenatal population.


Asunto(s)
Trastornos de la Coagulación Sanguínea Heredados/etnología , Polimorfismo Genético , Trombofilia/etnología , Trombofilia/genética , Alelos , Australia/epidemiología , Trastornos de la Coagulación Sanguínea Heredados/epidemiología , Factor V/genética , Femenino , Frecuencia de los Genes , Genotipo , Humanos , Metilenotetrahidrofolato Reductasa (NADPH2)/genética , Mutación , Embarazo , Prevalencia , Estudios Prospectivos , Protrombina/genética , Trombomodulina/genética , Trombofilia/epidemiología
14.
Gynecol Obstet Invest ; 54(1): 43-5, 2002.
Artículo en Inglés | MEDLINE | ID: mdl-12297717

RESUMEN

Thrombomodulin plays an important role in thromboresistance. A common mutation of the gene at amino acid 455 producing valine instead of alanine has been implicated as being a risk factor in myocardial infarction and chronic heart disease. However, its link with pre-eclampsia remains controversial. This report explores the Ala455Val point mutation in 280 women as a predictor of pre-eclampsia with the conclusion that in this regard, the dimorphism is essentially neutral.


Asunto(s)
Polimorfismo Genético/genética , Preeclampsia/genética , Trombomodulina/genética , Alelos , Australia , Secuencia de Bases , Estudios de Cohortes , ADN/química , ADN/genética , Femenino , Predisposición Genética a la Enfermedad , Humanos , Datos de Secuencia Molecular , Mutación/genética , Nueva Zelanda , Reacción en Cadena de la Polimerasa , Embarazo , Trombomodulina/química
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...