Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 3 de 3
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Clin Endocrinol Metab ; 98(12): 4953-60, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24106289

RESUMO

CONTEXT: Endothelial colony-forming cells (ECFCs) are the only putative endothelial progenitor cells capable of vasculogenesis, and their dysfunction may represent a risk factor for cardiovascular disease. Intrauterine growth restriction (IUGR) is a pregnancy-related disorder associated with long-term cardiovascular risk. OBJECTIVE: Our objective was to determine whether ECFCs derived from pregnancies complicated by IUGR exhibit altered vasculogenic potential. DESIGN AND SETTING: This was a prospective cohort study; patients were recruited at St. Mary's Hospital, Manchester, United Kingdom. PARTICIPANTS: Twenty-three women with normal pregnancies and 13 women with IUGR-complicated pregnancies at gestational ages above 37 weeks were included. MAIN OUTCOME MEASURES: Vasculogenic capacity of rigorously characterized ECFCs was investigated in vivo by measuring blood vessel formation in collagen/fibronectin gels implanted in mice; proliferative, migratory, and chemotactic abilities were assessed in cell culture. Placental uptake of fetal ECFCs, assessed by differences in arterial and venous cord blood content, was determined by flow cytometry. RESULTS: In vivo, IUGR ECFCs formed fewer blood vessels (P < .001) and capillaries (P = .001) compared with normal pregnancy-derived ECFCs. In culture conditions, IUGR ECFCs had reduced proliferation (P = .01) and migration (P = .007) and diminished chemotactic abilities to stromal cell-derived factor 1 (P = .007) coupled with reduced hypoxia-induced matrix metalloproteinase-2 release (P = .02). Finally, in IUGR pregnancies, the number of ECFCs was lower in arterial cord blood (P = .002) and placental uptake of cells was reduced (P < .001). CONCLUSIONS: ECFCs derived from IUGR cord blood are rarefied and dysfunctional, resulting in diminished vasculogenic potential; this could be a cause of placental dysfunction in IUGR, with long-term postnatal implications for cardiovascular function in offspring.


Assuntos
Endotélio Vascular/patologia , Retardo do Crescimento Fetal/patologia , Células-Tronco Fetais/patologia , Neovascularização Patológica/patologia , Capilares/enzimologia , Capilares/patologia , Capilares/fisiopatologia , Doenças Cardiovasculares/etiologia , Contagem de Células , Movimento Celular , Proliferação de Células , Células Cultivadas , Quimiotaxia , Estudos de Coortes , Regulação para Baixo , Endotélio Vascular/enzimologia , Endotélio Vascular/fisiopatologia , Feminino , Sangue Fetal , Retardo do Crescimento Fetal/sangue , Retardo do Crescimento Fetal/enzimologia , Retardo do Crescimento Fetal/fisiopatologia , Células-Tronco Fetais/enzimologia , Humanos , Metaloproteinase 2 da Matriz/metabolismo , Neovascularização Patológica/enzimologia , Neovascularização Patológica/fisiopatologia , Placenta/irrigação sanguínea , Placenta/patologia , Gravidez , Estudos Prospectivos
2.
Stem Cells ; 31(7): 1363-70, 2013 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-23554274

RESUMO

The potency of adult-derived circulating progenitor endothelial colony forming cells (ECFCs) is drastically surpassed by their fetal counterparts. Human pregnancy is associated with robust intensification of blood flow and vascular expansion in the uterus, crucial for placental perfusion and fetal supply. Here, we investigate whether fetal ECFCs transmigrate to maternal bloodstream and home to locations of maternal vasculogenesis, primarily the pregnant uterus. In the first instance, endothelial-like cells, originating from mouse fetuses expressing paternal eGFP, were identified within uterine endothelia. Subsequently, LacZ or enhanced green fluorescent protein (eGFP)-labeled human fetal ECFCs, transplanted into immunodeficient (NOD/SCID) fetuses on D15.5 pregnancy, showed similar integration into the mouse uterus by term. Mature endothelial controls (human umbilical vein endothelial cells), similarly introduced, were unequivocally absent. In humans, SRY was detected in 6 of 12 myometrial microvessels obtained from women delivering male babies. The copy number was calculated at 175 [IQR 149-471] fetal cells per millimeter square endothelium, constituting 12.5% of maternal vessel lumina. Cross-sections of similar human vessels, hybridized for Y-chromosome, positively identified endothelial-associated fetal cells. It appears that through ECFC donation, fetuses assist maternal uterine vascular expansion in pregnancy, potentiating placental perfusion and consequently their own fetal supply. In addition to fetal growth, this cellular mechanism holds implications for materno-fetal immune interactions and long-term maternal vascular health.


Assuntos
Células Endoteliais/fisiologia , Placenta/irrigação sanguínea , Gravidez/fisiologia , Útero/irrigação sanguínea , Animais , Diferenciação Celular/fisiologia , Células Cultivadas , Quimerismo , Feminino , Sangue Fetal , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos NOD , Camundongos SCID , Camundongos Transgênicos , Neovascularização Fisiológica/fisiologia , Placenta/metabolismo , Pré-Eclâmpsia/metabolismo , Células-Tronco , Útero/metabolismo
3.
Pregnancy Hypertens ; 1(1): 48-58, 2011 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26104231

RESUMO

The normal maternal cardiovascular adaptation to pregnancy involves a complex physiologic response to the growing conceptus, including alterations in maternal vascular endothelial cells that contribute to a profound fall in total systemic vascular resistance. There is a large body of evidence that adverse changes in the vascular endothelium underlie the multisystemic maternal manifestations of the hypertensive pregnancy disorder preeclampsia. Our knowledge is incomplete regarding the mechanisms of adaptive endothelial changes of normal pregnancy, and why these changes are attenuated or fail in women who develop preeclampsia. Endothelial progenitor cells (EPCs) are a heterogeneous population of cells that exist in both the fetus and adult. These cells can be mobilized into the circulation by growth factors and can then support the health of the vascular endothelium by several mechanisms. This review highlights some of the current understanding of EPCs, their potential role in pregnancy, and emerging evidence for EPC dysfunction in preeclampsia. We speculate that interference with nitric oxide (NO)-driven mobilization or activity of EPCs in the maternal circulation partially contributes to the widespread endothelial dysfunction underlying the clinical manifestations of preeclampsia. Potential roles of EPCs in the placenta and fetus are also considered.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...