Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Nat Commun ; 14(1): 1174, 2023 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-36859534

RESUMO

Placental abnormalities have been sporadically implicated as a source of developmental heart defects. Yet it remains unknown how often the placenta is at the root of congenital heart defects (CHDs), and what the cellular mechanisms are that underpin this connection. Here, we selected three mouse mutant lines, Atp11a, Smg9 and Ssr2, that presented with placental and heart defects in a recent phenotyping screen, resulting in embryonic lethality. To dissect phenotype causality, we generated embryo- and trophoblast-specific conditional knockouts for each of these lines. This was facilitated by the establishment of a new transgenic mouse, Sox2-Flp, that enables the efficient generation of trophoblast-specific conditional knockouts. We demonstrate a strictly trophoblast-driven cause of the CHD and embryonic lethality in one of the three lines (Atp11a) and a significant contribution of the placenta to the embryonic phenotypes in another line (Smg9). Importantly, our data reveal defects in the maternal blood-facing syncytiotrophoblast layer as a shared pathology in placentally induced CHD models. This study highlights the placenta as a significant source of developmental heart disorders, insights that will transform our understanding of the vast number of unexplained congenital heart defects.


Assuntos
Cardiopatias , Trofoblastos , Feminino , Gravidez , Animais , Camundongos , Placenta , Coração , Células Epiteliais , Camundongos Transgênicos
2.
PLoS One ; 15(1): e0226735, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-31917811

RESUMO

The major milestones in mouse placental development are well described, but our understanding is limited to how the placenta can adapt to damage or changes in the environment. By using stereology and expression of cell cycle markers, we found that the placenta grows under normal conditions not just by hyperplasia of trophoblast cells but also through extensive polyploidy and cell hypertrophy. In response to feeding a low protein diet to mothers prior to and during pregnancy, to mimic chronic malnutrition, we found that this normal program was altered and that it was influenced by the sex of the conceptus. Male fetuses showed intrauterine growth restriction (IUGR) by embryonic day (E) 18.5, just before term, whereas female fetuses showed IUGR as early as E16.5. This difference was correlated with differences in the size of the labyrinth layer of the placenta, the site of nutrient and gas exchange. Functional changes were implied based on up-regulation of nutrient transporter genes. The junctional zone was also affected, with a reduction in both glycogen trophoblast and spongiotrophoblast cells. These changes were associated with increased expression of Phlda2 and reduced expression of Egfr. Polyploidy, which results from endoreduplication, is a normal feature of trophoblast giant cells (TGC) but also spongiotrophoblast cells. Ploidy was increased in sinusoidal-TGCs and spongiotrophoblast cells, but not parietal-TGCs, in low protein placentas. These results indicate that the placenta undergoes a range of changes in development and function in response to poor maternal diet, many of which we interpret are aimed at mitigating the impacts on fetal and maternal health.


Assuntos
Aclimatação , Dieta com Restrição de Proteínas/efeitos adversos , Embrião de Mamíferos/citologia , Retardo do Crescimento Fetal/etiologia , Privação de Alimentos , Placenta/citologia , Animais , Proliferação de Células , Embrião de Mamíferos/fisiologia , Feminino , Desenvolvimento Fetal , Retardo do Crescimento Fetal/patologia , Células Gigantes , Masculino , Fenômenos Fisiológicos da Nutrição Materna , Camundongos , Camundongos Endogâmicos C57BL , Placenta/fisiologia , Gravidez , Trofoblastos/citologia , Trofoblastos/fisiologia
3.
Cell Res ; 28(8): 819-832, 2018 08.
Artigo em Inglês | MEDLINE | ID: mdl-30042384

RESUMO

The placenta is crucial for a successful pregnancy and the health of both the fetus and the pregnant woman. However, how the human trophoblast lineage is regulated, including the categorization of the placental cell subtypes is poorly understood. Here we performed single-cell RNA sequencing (RNA-seq) on sorted placental cells from first- and second-trimester human placentas. New subtypes of cells of the known cytotrophoblast cells (CTBs), extravillous trophoblast cells (EVTs), Hofbauer cells, and mesenchymal stromal cells were identified and cell-type-specific gene signatures were defined. Functionally, this study revealed many previously unknown functions of the human placenta. Notably, 102 polypeptide hormone genes were found to be expressed by various subtypes of placental cells, which suggests a complex and significant role of these hormones in regulating fetal growth and adaptations of maternal physiology to pregnancy. These results document human placental trophoblast differentiation at single-cell resolution and thus advance our understanding of human placentation during the early stage of pregnancy.


Assuntos
Hormônios Peptídicos/genética , Placenta/citologia , Placentação/genética , Trofoblastos/metabolismo , Sequência de Bases , Diferenciação Celular , Feminino , Expressão Gênica , Humanos , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/metabolismo , Gravidez , Primeiro Trimestre da Gravidez/genética , Primeiro Trimestre da Gravidez/metabolismo , Segundo Trimestre da Gravidez/genética , Segundo Trimestre da Gravidez/metabolismo , Análise de Sequência de RNA/métodos , Análise de Célula Única/métodos
4.
Sci Rep ; 7(1): 5575, 2017 07 17.
Artigo em Inglês | MEDLINE | ID: mdl-28717241

RESUMO

Trophoblast stem (TS) cells in the mouse derive from the polar trophectoderm of the blastocyst and persist through early gestation (to E8.5) to support placental development. Further development and growth is proposed to rely on layer-restricted progenitor cells. Stem cell antigen (Sca) -1 is a member of the Ly6 gene family and a known marker of stem cells in both hematopoietic and non-hematopoietic mouse tissues. Having identified that Sca-1 mRNA was highly expressed in mouse TS cells in culture, we found that it was also expressed in a subset of trophoblast within the chorion and labyrinth layer of the mouse placenta. Isolation and in vitro culture of Sca-1+ trophoblast cells from both differentiated TS cell cultures and dissected mouse placentae resulted in proliferating colonies that expressed known markers of TS cells. Furthermore, these cells could be stimulated to differentiate and expressed markers of both junctional zone and labyrinth trophoblast subtypes in a manner comparable to established mouse TS cell lines. Our results suggest that we have identified a subpopulation of TS cell-like cells that persist in the mid- to late- gestation mouse placenta as well as a cell surface protein that can be used to identify and isolate these cells.


Assuntos
Ataxina-1/genética , Ataxina-1/metabolismo , Células-Tronco Pluripotentes/citologia , Trofoblastos/citologia , Animais , Biomarcadores/metabolismo , Diferenciação Celular , Proliferação de Células , Células Cultivadas , Córion/citologia , Córion/metabolismo , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Idade Gestacional , Camundongos , Células-Tronco Pluripotentes/metabolismo , Gravidez , Trofoblastos/metabolismo , Regulação para Cima
5.
Circ Arrhythm Electrophysiol ; 8(2): 420-8, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25648353

RESUMO

BACKGROUND: N629D KCNH2 is a human missense long-QT2 mutation. Previously, we reported that the N629D/N629D mutation embryos disrupted cardiac looping, right ventricle development, and ablated IKr activity at E9.5. The present study evaluates the role of KCNH2 in vasculogenesis. METHODS AND RESULTS: N629D/N629D yolk sac vessels and aorta consist of sinusoids without normal arborization. Isolated E9.5 +/+ first branchial arches showed normal outgrowth of mouse ERG-positive/α-smooth muscle actin coimmunolocalized cells; however, outgrowth was grossly reduced in N629D/N629D. N629D/N629D aortas showed fewer α-smooth muscle actin positive cells that were not coimmunolocalized with mouse ERG cells. Transforming growth factor-ß treatment of isolated N629D/N629D embryoid bodies partially rescued this phenotype. Cultured N629D/N629D embryos recapitulate the same cardiovascular phenotypes as seen in vivo. Transforming growth factor-ß treatment significantly rescued these embryonic phenotypes. Both in vivo and in vitro, dofetilide treatment, over a narrow window of time, entirely recapitulated the N629D/N629D fetal phenotypes. Exogenous transforming growth factor-ß treatment also rescued the dofetilide-induced phenotype toward normal. CONCLUSIONS: Loss of function of KCNH2 mutations results in defects in cardiogenesis and vasculogenesis. Because many medications inadvertently block the KCNH2 potassium current, these novel findings seem to have clinical relevance.


Assuntos
Anormalidades Induzidas por Medicamentos/prevenção & controle , Células-Tronco Embrionárias/efeitos dos fármacos , Canais de Potássio Éter-A-Go-Go/antagonistas & inibidores , Canais de Potássio Éter-A-Go-Go/genética , Morte Fetal , Cardiopatias Congênitas/prevenção & controle , Mutação de Sentido Incorreto , Neovascularização Fisiológica/efeitos dos fármacos , Fenetilaminas/toxicidade , Bloqueadores dos Canais de Potássio/toxicidade , Sulfonamidas/toxicidade , Fator de Crescimento Transformador beta/farmacologia , Malformações Vasculares/prevenção & controle , Anormalidades Induzidas por Medicamentos/embriologia , Anormalidades Induzidas por Medicamentos/genética , Anormalidades Induzidas por Medicamentos/metabolismo , Animais , Células Cultivadas , Canal de Potássio ERG1 , Técnicas de Cultura Embrionária , Células-Tronco Embrionárias/metabolismo , Canais de Potássio Éter-A-Go-Go/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Genótipo , Cardiopatias Congênitas/induzido quimicamente , Cardiopatias Congênitas/embriologia , Cardiopatias Congênitas/genética , Cardiopatias Congênitas/metabolismo , Humanos , Camundongos da Linhagem 129 , Camundongos Transgênicos , Morfogênese/efeitos dos fármacos , Fenótipo , Transdução de Sinais , Malformações Vasculares/induzido quimicamente , Malformações Vasculares/embriologia , Malformações Vasculares/genética , Malformações Vasculares/metabolismo
6.
J Clin Invest ; 124(11): 4941-52, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25329693

RESUMO

There is strong evidence that overproduction of soluble fms-like tyrosine kinase-1 (sFLT1) in the placenta is a major cause of vascular dysfunction in preeclampsia through sFLT1-dependent antagonism of VEGF. However, the cause of placental sFLT1 upregulation is not known. Here we demonstrated that in women with preeclampsia, sFLT1 is upregulated in placental trophoblasts, while VEGF is upregulated in adjacent maternal decidual cells. In response to VEGF, expression of sFlt1 mRNA, but not full-length Flt1 mRNA, increased in cultured murine trophoblast stem cells. We developed a method for transgene expression specifically in mouse endometrium and found that endometrial-specific VEGF overexpression induced placental sFLT1 production and elevated sFLT1 levels in maternal serum. This led to pregnancy losses, placental vascular defects, and preeclampsia-like symptoms, including hypertension, proteinuria, and glomerular endotheliosis in the mother. Knockdown of placental sFlt1 with a trophoblast-specific transgene caused placental vascular changes that were consistent with excess VEGF activity. Moreover, sFlt1 knockdown in VEGF-overexpressing animals enhanced symptoms produced by VEGF overexpression alone. These findings indicate that sFLT1 plays an essential role in maintaining vascular integrity in the placenta by sequestering excess maternal VEGF and suggest that a local increase in VEGF can trigger placental overexpression of sFLT1, potentially contributing to the development of preeclampsia and other pregnancy complications.


Assuntos
Endométrio/enzimologia , Placenta/enzimologia , Pré-Eclâmpsia/enzimologia , Fator A de Crescimento do Endotélio Vascular/fisiologia , Receptor 1 de Fatores de Crescimento do Endotélio Vascular/metabolismo , Animais , Estudos de Casos e Controles , Indução Enzimática , Feminino , Expressão Gênica , Masculino , Camundongos , Gravidez , Receptor 1 de Fatores de Crescimento do Endotélio Vascular/genética
8.
Dev Biol ; 387(2): 131-41, 2014 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-24485853

RESUMO

The maternal vasculature within the placenta in primates and rodents is unique because it is lined by fetal cells of the trophoblast lineage and not by maternal endothelial cells. In addition to trophoblast cells that invade the uterine spiral arteries that bring blood into the placenta, other trophoblast subtypes sit at different levels of the vascular space. In mice, at least five distinct subtypes of trophoblast cells have been identified which engage maternal endothelial cells on the arterial and venous frontiers of the placenta, but which also form the channel-like spaces within it through a process analogous to formation of blood vessels (vasculogenic mimicry). These cells are all large, post-mitotic trophoblast giant cells. In addition to assuming endothelial cell-like characteristics (endothelial mimicry), they produce dozens of different hormones that are thought to regulate local and systemic maternal adaptations to pregnancy. Recent work has identified distinct molecular pathways in mice that regulate the morphogenesis of trophoblast cells on the arterial and venous sides of the vascular circuit that may be analogous to specification of arterial and venous endothelial cells.


Assuntos
Endotélio Vascular/citologia , Placenta/irrigação sanguínea , Placentação/fisiologia , Trofoblastos/citologia , Animais , Endotélio Vascular/patologia , Feminino , Humanos , Camundongos , Neoplasias/patologia , Gravidez , Trofoblastos/patologia
9.
Int J Dev Biol ; 54(2-3): 341-54, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-19876834

RESUMO

Trophoblast giant cells (TGCs) are the first cell type to terminally differentiate during embryogenesis and are of vital importance for implantation and modulation of post-implantation placentation. TGCs are mononuclear and polyploid but are heterogenous and dynamic. At least four different subtypes of TGCs are present within the mature placenta that have distinct cell lineage origins. The development of TGCs is complex and requires transition from the mitotic to the endoreduplication cell cycle and is regulated by a wide variety of factors. During early gestation, TGCs mediate blastocyst attachment and invasion into the uterine epithelium, regulate uterus decidualization, and anatomosis with maternal blood spaces to form the transient yolk sac placenta. During later gestation, TGCs secrete a wide array of hormones and paracrine factors, including steroid hormones and Prolactin-related cytokines, to target the maternal physiological systems for proper maternal adaptations to pregnancy and the fetal-maternal interface to ensure vasculature remodeling. The large number of mouse mutants with defects in TGC development and function are giving us significant new insights into the biology of these fascinating cells.


Assuntos
Células Gigantes/metabolismo , Placenta/metabolismo , Trofoblastos/metabolismo , Animais , Ciclo Celular , Diferenciação Celular , Feminino , Células Gigantes/citologia , Camundongos , Placenta/citologia , Gravidez , Trofoblastos/citologia
10.
Dev Dyn ; 238(10): 2564-74, 2009 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-19777589

RESUMO

The Mrj co-chaperone is expressed throughout the mouse conceptus, yet its requirement for placental development has prohibited a full understanding of its embryonic function. Here, we show that Mrj(-/-) embryos exhibit neural tube defects independent of the placenta phenotype, including exencephaly and thin-walled neural tubes. Molecular analyses revealed fewer proliferating cells and a down-regulation of early neural progenitor (Pax6, Olig2, Hes5) and neuronal (Nscl2, SCG10) cell markers in Mrj(-/-) neuroepithelial cells. Furthermore, Mrj(-/-) neurospheres are significantly smaller and form fewer secondary neurospheres indicating that Mrj is necessary for self-renewal of neural stem cells. However, the molecular function of Mrj in this context remains elusive because Mrj does not colocalize with Bmi-1, a self-renewal protein. Furthermore, unlike in Mrj(-/-) placentas, intermediate filament-containing aggregates do not accumulate in Mrj(-/-) neuroepithelium, ruling out nestin as a substrate for Mrj. Regardless, Mrj plays an important role in neural stem cell self-renewal.


Assuntos
Embrião de Mamíferos , Proteínas de Choque Térmico HSP40/metabolismo , Chaperonas Moleculares/metabolismo , Neurônios/fisiologia , Células-Tronco/fisiologia , Animais , Biomarcadores/metabolismo , Padronização Corporal/fisiologia , Diferenciação Celular/fisiologia , Proliferação de Células , Embrião de Mamíferos/anormalidades , Embrião de Mamíferos/anatomia & histologia , Embrião de Mamíferos/fisiologia , Epitélio/anatomia & histologia , Epitélio/fisiologia , Idade Gestacional , Proteínas de Choque Térmico HSP40/genética , Proteínas de Filamentos Intermediários/genética , Proteínas de Filamentos Intermediários/metabolismo , Camundongos , Camundongos Knockout , Chaperonas Moleculares/genética , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Nestina , Tubo Neural/anatomia & histologia , Tubo Neural/metabolismo , Neurônios/citologia , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Complexo Repressor Polycomb 1 , Proteínas Proto-Oncogênicas/genética , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Repressoras/genética , Proteínas Repressoras/metabolismo , Células-Tronco/citologia
11.
Dev Biol ; 335(1): 120-31, 2009 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-19716815

RESUMO

Prolonged maintenance of trophoblast stem (TS) cells requires fibroblast growth factor (FGF) 4 and embryonic fibroblast feeder cells or feeder cell-conditioned medium. Previous studies have shown that TGF-beta and Activin are sufficient to replace embryonic fibroblast-conditioned medium. Nodal, a member of the TGF-beta superfamily, is also known to be important in vivo for the maintenance of TS cells in the developing placenta. Our current studies indicate that TS cells do not express the Nodal co-receptor, Cripto, and do not respond directly to active Nodal in culture. Conversely, Activin subunits and their receptors are expressed in the placenta and TS cell cultures, with Activin predominantly expressed by trophoblast giant cells (TGCs). Differentiation of TS cells in the presence of TGC-conditioned medium or exogenous Activin results in a reduction in the expression of TGC markers. In line with TGC-produced Activin representing the active component in TGC-conditioned medium, this differentiation-inhibiting effect can be reversed by the addition of follistatin. Additional experiments in which TS cells were differentiated in the presence or absence of exogenous Activin or TGF-beta show that Activin but not TGF-beta results in the maintenance of expression of TS cell markers, prolongs the expression of syncytiotrophoblast markers, and significantly delays the expression of spongiotrophoblast and TGC markers. These results suggest that Activin rather than TGF-beta (or Nodal) acts directly on TS cells influencing both TS cell maintenance and cell fate, depending on whether the cells are also exposed to FGF4.


Assuntos
Ativinas/farmacologia , Diferenciação Celular/efeitos dos fármacos , Orelha Interna , Células-Tronco/efeitos dos fármacos , Células-Tronco/fisiologia , Trofoblastos , Receptores de Ativinas/genética , Receptores de Ativinas/metabolismo , Ativinas/genética , Ativinas/metabolismo , Animais , Biomarcadores/metabolismo , Diferenciação Celular/fisiologia , Linhagem da Célula , Células Cultivadas , Meios de Cultivo Condicionados/química , Orelha Interna/citologia , Orelha Interna/embriologia , Embrião de Mamíferos/anatomia & histologia , Embrião de Mamíferos/metabolismo , Fator de Crescimento Epidérmico/genética , Fator de Crescimento Epidérmico/metabolismo , Feminino , Fator 4 de Crescimento de Fibroblastos/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Inibinas/genética , Inibinas/metabolismo , Masculino , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/metabolismo , Camundongos , Análise em Microsséries , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Proteína Nodal/genética , Proteína Nodal/metabolismo , Proteína Nodal/farmacologia , Comunicação Parácrina/fisiologia , Placenta/citologia , Placenta/metabolismo , Gravidez , Células-Tronco/citologia , Fator de Crescimento Transformador beta/farmacologia , Trofoblastos/citologia , Trofoblastos/efeitos dos fármacos
12.
Circ Res ; 103(12): 1483-91, 2008 Dec 05.
Artigo em Inglês | MEDLINE | ID: mdl-18948620

RESUMO

Loss-of-function mutations in the human ERG1 potassium channel (hERG1) frequently underlie the long QT2 (LQT2) syndrome. The role of the ERG potassium channel in cardiac development was elaborated in an in vivo model of a homozygous, loss-of-function LQT2 syndrome mutation. The hERG N629D mutation was introduced into the orthologous mouse gene, mERG, by homologous recombination in mouse embryonic stem cells. Intact homozygous embryos showed abrupt cessation of the heart beat. N629D/N629D embryos die in utero by embryonic day 11.5. Their developmental defects include altered looping architecture, poorly developed bulbus cordis, and distorted aortic sac and branchial arches. N629D/N629D myocytes from embryonic day 9.5 embryos manifested complete loss of I(Kr) function, depolarized resting potential, prolonged action potential duration (LQT), failure to repolarize, and propensity to oscillatory arrhythmias. N629D/N629D myocytes manifest calcium oscillations and increased sarcoplasmic reticulum Ca(+2) content. Although the N629D/N629D protein is synthesized, it is mainly located intracellularly, whereas +/+ mERG protein is mainly in plasmalemma. N629D/N629D embryos show robust apoptosis in craniofacial regions, particularly in the first branchial arch and, to a lesser extent, in the cardiac outflow tract. Because deletion of Hand2 produces apoptosis, in similar regions and with a similar final developmental phenotype, Hand2 expression was evaluated. Robust decrease in Hand2 expression was observed in the secondary heart field in N629D/N629D embryos. In conclusion, loss of I(Kr) function in N629D/N629D cardiovascular system leads to defects in cardiac ontogeny in the first branchial arch, outflow tract, and the right ventricle.


Assuntos
Canais de Potássio Éter-A-Go-Go/genética , Regulação da Expressão Gênica no Desenvolvimento/genética , Homozigoto , Mutação de Sentido Incorreto/genética , Disfunção Ventricular Direita/embriologia , Disfunção Ventricular Direita/genética , Substituição de Aminoácidos/genética , Animais , Asparagina/genética , Ácido Aspártico/genética , Débito Cardíaco/genética , Canal de Potássio ERG1 , Feminino , Ventrículos do Coração/embriologia , Ventrículos do Coração/metabolismo , Síndrome do QT Longo/embriologia , Síndrome do QT Longo/genética , Síndrome do QT Longo/mortalidade , Camundongos , Camundongos Mutantes , Gravidez , Disfunção Ventricular Direita/mortalidade
13.
Development ; 135(19): 3311-20, 2008 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-18776147

RESUMO

Trophoblast giant cells are instrumental in promoting blood flow towards the mouse embryo by invading the uterine endometrium and remodelling the maternal vasculature. This process involves the degradation of the perivascular smooth muscle layer and the displacement of vascular endothelial cells to form trophoblast-lined blood sinuses. How this vascular remodelling is achieved at the molecular level remains largely elusive. Here, we show that two placenta-specific cathepsins, Cts7 and Cts8, are expressed in distinct but largely overlapping subsets of giant cells that are in direct contact with maternal arteries. We find that Cts8, but not Cts7, has the capacity to mediate loss of smooth muscle alpha-actin and to disintegrate blood vessels. Consequently, conditional ubiquitous overexpression of Cts8 leads to midgestational embryonic lethality caused by severe vascularization defects. In addition, both cathepsins determine trophoblast cell fate by inhibiting the self-renewing capacity of trophoblast stem cells when overexpressed in vitro. Similarly, transgenic overexpression of Cts7 and Cts8 affects trophoblast proliferation and differentiation by prolonging mitotic cell cycle progression and promoting giant cell differentiation, respectively. We also show that the cell cycle effect is directly caused by some proportion of CTS7 localizing to the nucleus, highlighting the emerging functional diversity of these typically lysosomal proteases in distinct intracellular compartments. Our findings provide evidence for the highly specialized functions of closely related cysteine cathepsin proteases in extra-embryonic development, and reinforce their importance for a successful outcome of pregnancy.


Assuntos
Catepsinas/fisiologia , Placenta/irrigação sanguínea , Placenta/enzimologia , Trofoblastos/fisiologia , Animais , Catepsinas/genética , Ciclo Celular , Diferenciação Celular , Proliferação de Células , Desenvolvimento Embrionário/genética , Endossomos/enzimologia , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Regulação Enzimológica da Expressão Gênica , Células Gigantes/citologia , Células Gigantes/enzimologia , Lisossomos/enzimologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos CBA , Camundongos Transgênicos , Modelos Biológicos , Gravidez , Proteínas/genética , Proteínas/fisiologia , Trofoblastos/citologia
14.
Mol Genet Metab ; 91(1): 85-97, 2007 May.
Artigo em Inglês | MEDLINE | ID: mdl-17369066

RESUMO

Hyperhomocyst(e)inemia is a metabolic derangement that is linked to the distribution of folate pools, which provide one-carbon units for biosynthesis of purines and thymidylate and for remethylation of homocysteine to form methionine. In humans, methionine synthase deficiency results in the accumulation of methyltetrahydrofolate at the expense of folate derivatives required for purine and thymidylate biosynthesis. Complete ablation of methionine synthase activity in mice results in embryonic lethality. Other mouse models for hyperhomocyst(e)inemia have normal or reduced levels of methyltetrahydrofolate and are not embryonic lethal, although they have decreased ratios of AdoMet/AdoHcy and impaired methylation. We have constructed a mouse model with a gene trap insertion in the Mtrr gene specifying methionine synthase reductase, an enzyme essential for the activity of methionine synthase. This model is a hypomorph, with reduced methionine synthase reductase activity, thus avoiding the lethality associated with the absence of methionine synthase activity. Mtrr(gt/gt) mice have increased plasma homocyst(e)ine, decreased plasma methionine, and increased tissue methyltetrahydrofolate. Unexpectedly, Mtrr(gt/gt) mice do not show decreases in the AdoMet/AdoHcy ratio in most tissues. The different metabolite profiles in the various genetic mouse models for hyperhomocyst(e)inemia may be useful in understanding biological effects of elevated homocyst(e)ine.


Assuntos
Ferredoxina-NADP Redutase/genética , Ferredoxina-NADP Redutase/metabolismo , Ácido Fólico/metabolismo , Metionina/metabolismo , Animais , Encéfalo/embriologia , Encéfalo/metabolismo , Modelos Animais de Doenças , Embrião de Mamíferos/metabolismo , Feminino , Coração/embriologia , Homocisteína/sangue , Homocisteína/metabolismo , Hiper-Homocisteinemia/enzimologia , Hiper-Homocisteinemia/metabolismo , Rim/embriologia , Rim/metabolismo , Fígado/embriologia , Fígado/metabolismo , Masculino , Metionina/sangue , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , RNA Mensageiro/metabolismo , S-Adenosil-Homocisteína/metabolismo , S-Adenosilmetionina/metabolismo
15.
Dev Biol ; 304(2): 567-78, 2007 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-17289015

RESUMO

Trophoblast giant cells (TGCs) are the first terminally differentiated subtype to form in the trophoblast cell lineage in rodents. In addition to mediating implantation, they are the main endocrine cells of the placenta, producing several hormones which regulate the maternal endocrine and immune systems and promote maternal blood flow to the implantation site. Generally considered a homogeneous population, TGCs have been identified by their expression of genes encoding placental lactogen 1 or proliferin. In the present study, we have identified a number of TGC subtypes, based on morphology and molecular criteria and demonstrated a previously underappreciated diversity of TGCs. In addition to TGCs that surround the implantation site and form the interface with the maternal deciduas, we demonstrate at least three other unique TGC subtypes: spiral artery-associated TGCs, maternal blood canal-associated TGCs and a TGC within the sinusoidal spaces of the labyrinth layer of the placenta. All four TGC subtypes could be identified based on the expression patterns of four genes: Pl1, Pl2, Plf (encoded by genes of the prolactin/prolactin-like protein/placental lactogen gene locus), and Ctsq (from a placental-specific cathepsin gene locus). Each of these subtypes was detected in differentiated trophoblast stem cell cultures and can be differentially regulated; treatment with retinoic acid induces Pl1/Plf+ TGCs preferentially. Furthermore, cell lineage tracing studies indicated unique origins for different TGC subtypes, in contrast with previous suggestions that secondary TGCs all arise from Tpbpa+ ectoplacental cone precursors.


Assuntos
Células Gigantes/citologia , Placenta/citologia , Trofoblastos/citologia , Animais , Catepsinas/biossíntese , Catepsinas/genética , Diferenciação Celular , Linhagem da Célula , Feminino , Células Gigantes/metabolismo , Camundongos , Placenta/metabolismo , Lactogênio Placentário/biossíntese , Lactogênio Placentário/genética , Poliploidia , Gravidez , Trofoblastos/metabolismo
16.
Genes Dev ; 21(1): 85-97, 2007 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-17210791

RESUMO

The inactivation of the retinoblastoma (Rb) tumor suppressor gene in mice results in ectopic proliferation, apoptosis, and impaired differentiation in extraembryonic, neural, and erythroid lineages, culminating in fetal death by embryonic day 15.5 (E15.5). Here we show that the specific loss of Rb in trophoblast stem (TS) cells, but not in trophoblast derivatives, leads to an overexpansion of trophoblasts, a disruption of placental architecture, and fetal death by E15.5. Despite profound placental abnormalities, fetal tissues appeared remarkably normal, suggesting that the full manifestation of fetal phenotypes requires the loss of Rb in both extraembryonic and fetal tissues. Loss of Rb resulted in an increase of E2f3 expression, and the combined ablation of Rb and E2f3 significantly suppressed Rb mutant phenotypes. This rescue appears to be cell autonomous since the inactivation of Rb and E2f3 in TS cells restored placental development and extended the life of embryos to E17.5. Taken together, these results demonstrate that loss of Rb in TS cells is the defining event causing lethality of Rb(-/-) embryos and reveal the convergence of extraembryonic and fetal functions of Rb in neural and erythroid development. We conclude that the Rb pathway plays a critical role in the maintenance of a mammalian stem cell population.


Assuntos
Placenta/metabolismo , Proteína do Retinoblastoma/fisiologia , Células-Tronco/citologia , Trofoblastos/citologia , Animais , Apoptose , Diferenciação Celular , Fator de Transcrição E2F3/genética , Fator de Transcrição E2F3/metabolismo , Perda do Embrião/metabolismo , Perda do Embrião/patologia , Embrião de Mamíferos/citologia , Embrião de Mamíferos/metabolismo , Feto/citologia , Feto/embriologia , Feto/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Mutação/genética , Fenótipo , Placenta/anormalidades , Placenta/citologia , Proteína do Retinoblastoma/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Células-Tronco/metabolismo , Trofoblastos/metabolismo
17.
Dev Biol ; 294(2): 445-56, 2006 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-16616120

RESUMO

The uterus undergoes a series of dramatic changes in response to an implanting conceptus that, in some mammalian species, includes differentiation of the endometrial stroma into decidual tissue. This process, called decidualization, can be induced artificially in rodents indicating that the conceptus may not be essential for a proper maternal response in early pregnancy. In order to test this hypothesis, we determined if and how the conceptus affects uterine gene expression. We identified 5 genes (Angpt1, Angpt2, Dtprp, G1p2 and Prlpa) whose steady-state levels in the uterus undergoing decidualization depends on the presence of a conceptus. In situ hybridization revealed region-specific effects which suggested that various components of the conceptus and more than one signal from the conceptus are likely responsible for altering decidual cell function. Using cell culture models we found that trophoblast giant cells secrete a type I interferon-like molecule which can induce G1p2 expression in endometrial stromal cells. Finally, decidual Prlpa expression was reduced in the uterus adjacent to Hand1- and Ets2-deficient embryos, suggesting that normal trophoblast giant cells in the placenta are required for the conceptus-dependent effects on Prlpa expression in the mesometrial decidua. Overall, these results provide support for the hypothesis that molecular signals from the mouse conceptus have local effects on uterine gene expression during decidualization.


Assuntos
Decídua/fisiologia , Embrião de Mamíferos/fisiologia , Endométrio , Comunicação Parácrina , Proteína 2 Semelhante a Angiopoietina , Proteínas Semelhantes a Angiopoietina , Angiopoietinas , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Proteínas Sanguíneas/genética , Proteínas Sanguíneas/metabolismo , Células Cultivadas , Citocinas/genética , Citocinas/metabolismo , Decídua/citologia , Deciduoma/citologia , Deciduoma/metabolismo , Embrião de Mamíferos/citologia , Endométrio/citologia , Endométrio/fisiologia , Feminino , Perfilação da Expressão Gênica , Regulação da Expressão Gênica no Desenvolvimento , Humanos , Hibridização In Situ , Peptídeos e Proteínas de Sinalização Intercelular , Interferons/genética , Interferons/metabolismo , Masculino , Camundongos , Análise de Sequência com Séries de Oligonucleotídeos , Gravidez , Proteínas da Gravidez/genética , Proteínas da Gravidez/metabolismo , Prolactina/análogos & derivados , Prolactina/genética , Prolactina/metabolismo , Proteína Proto-Oncogênica c-ets-2/genética , Proteína Proto-Oncogênica c-ets-2/metabolismo , Ubiquitinas/genética , Ubiquitinas/metabolismo
18.
Dev Biol ; 284(1): 12-24, 2005 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-15963972

RESUMO

Cells of the trophoblast lineage make up the epithelial compartment of the placenta, and their rapid development is essential for the establishment and maintenance of pregnancy. A diverse array of specialized trophoblast subtypes form throughout gestation and are responsible for mediating implantation, as well as promotion of blood to the implantation site, changes in maternal physiology, and nutrient and gas exchange between the fetal and maternal blood supplies. Within the last decade, targeted mutations in mice and the study of trophoblast stem cells in vitro have contributed greatly to our understanding of trophoblast lineage development. Here, we review recent insights into the molecular pathways regulating trophoblast lineage segregation, stem cell maintenance, and subtype differentiation.


Assuntos
Diferenciação Celular/fisiologia , Linhagem da Célula/fisiologia , Modelos Biológicos , Placenta/embriologia , Transdução de Sinais/fisiologia , Células-Tronco/fisiologia , Trofoblastos/fisiologia , Animais , Fator 4 de Crescimento de Fibroblastos/metabolismo , Camundongos , Placenta/citologia
19.
Dev Biol ; 271(1): 26-37, 2004 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-15196947

RESUMO

The trophoblast cell lineage is an interesting model system because it is composed of a limited number of cell types that are spatially patterned. Trophoblast stem (TS) cells reside within a layer called the chorion and either remain as stem cells or differentiate into spongiotrophoblast (SpT), trophoblast giant (TG) cells or syncytiotrophoblast cells (SynT) of the labyrinth. Maintenance of the TS phenotype is dependent on stimulation by FGF4, whereas differentiation and/or maintenance of the differentiated derivatives are dependent on key transcription factors: Mash2 for SpT, Hand1 for TG cells and Gcm1 for SynT cells. TS cells proliferate and retain their stem cell phenotype in culture in response to FGF4 and an additional factor(s) that can be provided by conditioned medium from embryonic fibroblast feeder cells (CM). To understand the functions of Hand1, Mash2 and Gcm1 at a cellular level, we tested the effects of their ectopic and over-expression on the ability of TS cells to either continue to proliferate or differentiate into their alternative fates. Expression of Mash2 alone had no effects on TS cell differentiation. However, Mash2-transfected cells continued to divide longer after withdrawal of FGF/CM. Hand1 promoted TGC differentiation, even in the continued presence of FGF4/CM. Stra13, another bHLH factor gene that is expressed in TG cells, also induced TG differentiation. Gcm1 induced a rapid arrest of TS proliferation but, in contrast to Hand1 and Stra13, blocked TG cell differentiation. Although Gcm1 was not sufficient to promote SynT formation, expression of an antisense Gcm1 transcript blocked SynT differentiation. These data suggest that Mash2 functions to promote transient FGF4-independent amplification of trophoblast cells that are progressing towards the SpT and TG cell phenotype. By contrast, Hand1 and Stra13 promote cell cycle exit and restrict cells towards the TG fate, whereas Gcm1 promotes cell cycle exit and restriction towards the SynT fate.


Assuntos
Diferenciação Celular/fisiologia , Proteínas de Ligação a DNA/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Proteínas de Homeodomínio/metabolismo , Neuropeptídeos/metabolismo , Transdução de Sinais/fisiologia , Fatores de Transcrição/metabolismo , Trofoblastos/fisiologia , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos , Northern Blotting , Western Blotting , Proteínas de Ciclo Celular/metabolismo , Linhagem Celular , DNA Complementar/genética , Proteínas de Ligação a DNA/genética , Densitometria , Fator 4 de Crescimento de Fibroblastos , Fatores de Crescimento de Fibroblastos/metabolismo , Proteínas de Homeodomínio/genética , Imuno-Histoquímica , Hibridização In Situ , Camundongos , Neuropeptídeos/genética , Proteínas Nucleares , Plasmídeos/genética , Proteínas Proto-Oncogênicas/metabolismo , Transdução de Sinais/genética , Fatores de Transcrição/genética , Transfecção , Trofoblastos/metabolismo
20.
Cardiovasc Res ; 61(2): 268-77, 2004 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-14736543

RESUMO

OBJECTIVE: The long QT syndrome, N629D HERG mutation, alters the pore selectivity signature sequence, GFGN to GFGD. Heterologous co-expression of N629D and the wildtype HERG resulted in a relative loss of the selectivity of K+ over Na+, but its physiologic relevance has not been assessed in cardiac myocytes. METHODS AND RESULTS: Accordingly, N629D was overexpressed, via adenoviral gene transfer, in cardiomyocytes derived from mouse stem cells. Three IKr phenotypes were observed: (1) the wildtype-like IKr showed inward rectification and a positive tail current; (2) the N629D-like IKr showed outward rectification and an inward tail current; and (3) intermediate IKr showed a small outward tail current. Action potentials (AP) were paired with the IKr measurements in each cell. Resting membrane potential (RMP) was critically dependent on the IKr phenotype. The resting membrane potential of the cells was -61 +/- 5 mV (n=40) in wildtype, -63 +/- 3 mV (n=18) in wildtype-like IKr phenotype, -30 +/- 2 mV (n=12) in N629D-like and -47 +/- 2 mV (n=24) in intermediate phenotype (p<0.00001). Triggered action potential durations (APD) were: 62 +/- 12 ms (n=6) in wildtype, 65 +/- 11 ms (n=6) in wildtype-like IKr phenotypes and 106 +/- 10 ms (n=6) (p<0.01) in intermediate IKr phenotypes. Lowering [K+]o hyperpolarized wildtype cells and cells with a wildtype-like IKr phenotype, but depolarized those with intermediate phenotype (from -45 +/- 1 to -35 +/- 0.5 mV (n=12), p<0.01). In 6 of 12 cells, with intermediate phenotype, the hypokalemia-induced depolarization resulted in triggered activity. TTX suppressed this triggered activity. CONCLUSION: Overexpression of N629D in cardiomyocytes derived from stem cells results in phenotypic variability in IKr, which was the critical determinant of the resting membrane potential, action potential duration and arrhythmogenic response to low [K+]o.


Assuntos
Proteínas de Transporte de Cátions/genética , Síndrome do QT Longo/metabolismo , Miócitos Cardíacos/metabolismo , Mutação Puntual , Canais de Potássio de Abertura Dependente da Tensão da Membrana , Canais de Potássio/genética , Potenciais de Ação , Adenoviridae/genética , Animais , Diferenciação Celular , Células Cultivadas , Canais de Potássio Éter-A-Go-Go , Vetores Genéticos/farmacologia , Proteínas de Fluorescência Verde , Proteínas Luminescentes/genética , Potenciais da Membrana , Camundongos , Técnicas de Patch-Clamp , Canais de Potássio/metabolismo , Células-Tronco/citologia , Transdução Genética/métodos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA