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











Base de dados
Intervalo de ano de publicação
1.
Clin Chim Acta ; 474: 137-146, 2017 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-28935154

RESUMO

Syncytin-1, a human endogenous retroviral envelope gene (HERVW1), is specifically expressed in placental trophoblasts and mediates the formation of syncytiotrophoblasts through a fusogenic activity. Syncytin-1 expression deficiency has been repeatedly observed in preeclamptic/IUGR placentas. Previous gene knockout studies indicated that in mice, complete syncytin-A null mouse embryos died in utero between 11.5 and 13.5days of gestation. However, the complete knockout model could not fully recapitulate the mid- to third-trimester, time-specific syncytin-1 deficiency in preeclampsia patients. To construct a preeclampsia model and to better investigate the function of syncytin in placental development, we created a mouse inducible knockout model of syncytin-1A gene. It was found that the disruption of syncytin-A at E11.5 to E17.5 is associated with significant morphological changes in placentas and fetuses. Moreover, syncytin-A disruption led to extensive vasculature abnormalities in the labyrinth, with irregular distribution and reduced number of fetal microvessels. Moreover, Syncytin-A knockout affected neovascularization-related gene expression in labyrinth and the maternal plasma level of sVEGFR-1, and a dramatic increase of sFlt-1/PlGF ratio. These findings indicate that syncytin-A may be involved in the placenta angiogenesis and potentially, the development of preeclampsia. The new model could be a useful tool for studying the pathogenesis and management of preeclampsia.


Assuntos
Produtos do Gene env/deficiência , Produtos do Gene env/genética , Placenta/irrigação sanguínea , Pré-Eclâmpsia/genética , Pré-Eclâmpsia/fisiopatologia , Proteínas da Gravidez/deficiência , Proteínas da Gravidez/genética , Animais , Feminino , Técnicas de Inativação de Genes , Camundongos , Camundongos Endogâmicos C57BL , Mães , Gravidez , Receptor 1 de Fatores de Crescimento do Endotélio Vascular/sangue
2.
Methods Mol Biol ; 1658: 133-145, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28861788

RESUMO

Traditional primary and secondary cell cultures have been used for the investigation of prion biology and disease for many years. While both types of cultures produce highly valid and immensely valuable results, they also have their limitations; traditional cell lines are often derived from cancers, therefore subject to numerous DNA changes, and primary cultures are labor-intensive and expensive to produce requiring sacrifice of many animals. Neural stem cell (NSC) cultures are a relatively new technology to be used for the study of prion biology and disease. While NSCs are subject to their own limitations-they are generally cultured ex vivo in environments that artificially force their growth-they also have their own unique advantages. NSCs retain the ability for self-renewal and can therefore be propagated in culture similarly to secondary cultures without genetic manipulation. In addition, NSCs are multipotent; they can be induced to differentiate into mature cells of central nervous system (CNS) linage. The combination of self-renewal and multipotency allows NSCs to be used as a primary cell line over multiple generations saving time, costs, and animal harvests, thus providing a valuable addition to the existing cell culture repertoire used for investigation of prion biology and disease. Furthermore, NSC cultures can be generated from mice of any genotype, either by embryonic harvest or harvest from adult brain, allowing gene expression to be studied without further genetic manipulation. This chapter describes a standard method of culturing adult NSCs and assays for monitoring NSC growth, migration, and differentiation and revisits basic reactive oxygen species detection in the context of NSC cultures.


Assuntos
Células-Tronco Adultas/metabolismo , Bioensaio , Células-Tronco Neurais/metabolismo , Neurônios/metabolismo , Proteínas da Gravidez/genética , Espécies Reativas de Oxigênio/metabolismo , Esferoides Celulares/metabolismo , Células-Tronco Adultas/citologia , Animais , Diferenciação Celular , Movimento Celular , Proliferação de Células , Fluoresceínas/química , Corantes Fluorescentes/química , Expressão Gênica , Técnicas de Inativação de Genes , Camundongos , Células-Tronco Neurais/citologia , Neurônios/citologia , Proteínas da Gravidez/deficiência , Cultura Primária de Células , Esferoides Celulares/citologia
3.
Mol Hum Reprod ; 22(2): 130-42, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26646502

RESUMO

STUDY HYPOTHESIS: Placental growth factor (PGF) is expressed in the developing mouse brain and contributes to vascularization and vessel patterning. STUDY FINDING: PGF is dynamically expressed in fetal mouse brain, particularly forebrain, and is essential for normal cerebrovascular development. WHAT IS KNOWN ALREADY: PGF rises in maternal plasma over normal human and mouse pregnancy but is low in many women with the acute onset hypertensive syndrome, pre-eclampsia (PE). Little is known about the expression of PGF in the fetus during PE. Pgf  (-/-) mice appear normal but recently cerebral vascular defects were documented in adult Pgf  (-/-) mice. STUDY DESIGN, SAMPLES/MATERIALS, METHODS: Here, temporal-spatial expression of PGF is mapped in normal fetal mouse brains and cerebral vasculature development is compared between normal and congenic Pgf  (-/-) fetuses to assess the actions of PGF during cerebrovascular development. Pgf/PGF, Vegfa/VEGF, Vegf receptor (Vegfr)1 and Vegfr2 expression were examined in the brains of embryonic day (E)12.5, 14.5, 16.5 and 18.5 C57BL/6 (B6) mice using quantitative PCR and immunohistochemistry. The cerebral vasculature was compared between Pgf  (-/-) and B6 embryonic and adult brains using whole mount techniques. Vulnerability to cerebral ischemia was investigated using a left common carotid ligation assay. MAIN RESULTS AND THE ROLE OF CHANCE: Pgf/PGF and Vegfr1 are highly expressed in E12.5-14.5 forebrain relative to VEGF and Vegfr2. Vegfa/VEGF is relatively more abundant in hindbrain (HB). PGF and VEGF expression were similar in midbrain. Delayed HB vascularization was seen at E10.5 and 11.5 in Pgf  (-/-) brains. At E14.5, Pgf  (-/-) circle of Willis showed unilateral hypoplasia and fewer collateral vessels, defects that persisted post-natally. Functionally, adult Pgf  (-/-) mice experienced cerebral ischemia after left common carotid arterial occlusion while B6 mice did not. LIMITATIONS, REASONS FOR CAUTION: Since Pgf  (-/-) mice were used, consequences of complete absence of maternal and fetal PGF were defined. Therefore, the effects of maternal versus fetal PGF deficiency on cerebrovascular development cannot be separated. However, as PGF was strongly expressed in the developing brain at all timepoints, we suggest that local PGF has a more important role than distant maternal or placental sources. Full PGF loss is not expected in PE pregnancies, predicting that the effects of PGF deficiency identified in this model will be more severe than any effects in PE-offspring. WIDER IMPLICATIONS OF THE FINDINGS: These studies provoke the question of whether PGF expression is decreased and cerebral vascular maldevelopment occurs in fetuses who experience a preeclamptic gestation. These individuals have already been reported to have elevated risk for stroke and cognitive impairments. LARGE SCALE DATA: N/A. STUDY FUNDING AND COMPETING INTERESTS: This work was supported by awards from the Natural Sciences and Engineering Research Council, the Canada Research Chairs Program and the Canadian Foundation for Innovation to B.A.C. and by training awards from the Universidade Federal de Pernambuco and Conselho Nacional de Desenvolvimento Cientifico e Tecnologico (CNPq), Brazil to R.L.L.; Queen's University to V.R.K. and the Canadian Institutes of Health Research to M.T.R. The work of P.C. is supported by the Belgian Science Policy BELSPO-IUAP7/03, Structural funding by the Flemish Government-Methusalem funding, and the Flemish Science Fund-FWO grants. There were no competing interests.


Assuntos
Isquemia Encefálica/genética , Encéfalo/metabolismo , Estenose Coronária/genética , Neovascularização Patológica/genética , Proteínas da Gravidez/deficiência , Animais , Encéfalo/irrigação sanguínea , Encéfalo/patologia , Isquemia Encefálica/metabolismo , Isquemia Encefálica/patologia , Artéria Carótida Primitiva/metabolismo , Artéria Carótida Primitiva/patologia , Estenose Coronária/metabolismo , Estenose Coronária/patologia , Modelos Animais de Doenças , Embrião de Mamíferos , Feminino , Feto , Regulação da Expressão Gênica , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neovascularização Patológica/metabolismo , Neovascularização Patológica/patologia , Fator de Crescimento Placentário , Pré-Eclâmpsia/genética , Pré-Eclâmpsia/metabolismo , Pré-Eclâmpsia/patologia , Gravidez , Proteínas da Gravidez/genética , Transdução de Sinais , Fator A de Crescimento do Endotélio Vascular/genética , Fator A de Crescimento do Endotélio Vascular/metabolismo , Receptor 1 de Fatores de Crescimento do Endotélio Vascular/genética , Receptor 1 de Fatores de Crescimento do Endotélio Vascular/metabolismo , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/genética , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo
4.
Reproduction ; 149(2): R91-102, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25342175

RESUMO

Mammalian pregnancy involves tremendous de novo maternal vascular construction to adequately support conceptus development. In early mouse decidua basalis (DB), maternal uterine natural killer (uNK) cells oversee this process directing various aspects during the formation of supportive vascular networks. The uNK cells recruited to early implantation site DB secrete numerous factors that act in the construction of early decidual vessels (neoangiogenesis) as well as in the alteration of the structural components of newly developing and existing vessels (pruning and remodeling). Although decidual and placental development sufficient to support live births occur in the absence of normally functioning uNK cells, development and structure of implantation site are optimized through the presence of normally activated uNK cells. Human NK cells are also recruited to early decidua. Gestational complications including recurrent spontaneous abortion, fetal growth restriction, preeclampsia, and preterm labor are linked with the absence of human NK cell activation via paternally inherited conceptus transplantation antigens. This review summarizes the roles that mouse uNK cells normally play in decidual neoangiogenesis and spiral artery remodeling in mouse pregnancy and briefly discusses changes in early developmental angiogenesis due to placental growth factor deficiency.


Assuntos
Decídua/irrigação sanguínea , Células Matadoras Naturais/fisiologia , Útero/citologia , Aborto Habitual , Aborto Animal , Animais , Feminino , Retardo do Crescimento Fetal , Humanos , Camundongos , Neovascularização Fisiológica , Trabalho de Parto Prematuro , Placenta/irrigação sanguínea , Fator de Crescimento Placentário , Pré-Eclâmpsia , Gravidez , Proteínas da Gravidez/deficiência , Proteínas da Gravidez/fisiologia
5.
Placenta ; 35(9): 772-5, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-25106880

RESUMO

Effects of placental growth factor (PGF), an angiokine product of fetal trophoblasts and maternal decidual cells, on early decidual angiogenesis are undefined. We used whole-mount immunofluorescence analyses to compare uterus and gestation day 4.5-9.5 mouse implantation sites that differed genetically in fetal or maternal PGF deficiency. Implant site number and embryonic development were similar in Pgf(-/-) and Pgf(+/+) females although Pgf(-/-) lymphatic vessels were anomalous. Correct, fine branching angiogenesis of anti-mesometrial vessels required both conceptus and maternal PGF; correct mesometrial branching angiogenesis depended solely upon conceptus PGF. Thus, PGF is non-redundant for optimizing branching angiogenesis in early decidua.


Assuntos
Implantação do Embrião , Neovascularização Fisiológica , Proteínas da Gravidez/deficiência , Útero/irrigação sanguínea , Animais , Feminino , Masculino , Camundongos , Fator de Crescimento Placentário , Gravidez
6.
FASEB J ; 28(8): 3703-19, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24812088

RESUMO

Exosomes are extracellular vesicles that mediate intercellular communication and are involved in several biological processes. The objective of our study was to determine whether endogenous retrovirus group WE, member l (ERVWE1)/syncytin-1 and endogenous retrovirus group FRD, member 1 (ERVFRDE1)/syncytin-2, encoded by human endogenous retrovirus (HERV) envelope (env) genes, are present at the surface of exosomes produced by placenta-derived villous cytotrophoblasts and whether they play a role in cellular uptake of exosomes. In addition, we sought to determine whether these proteins are present in various abundances in serum-derived exosomes from normal pregnant women vs. women with preeclampsia (PE). Isolated exosomes were analyzed for their content by Western blot, a bead-associated flow cytometry approach, and a syncytin-2 ELISA. Binding and uptake were tested through confocal and electron microscopy using the BeWo choriocarcinoma cell line. Quality control of exosome preparations consisted of detection of exosomal and nonexosomal markers. Exosome-cell interactions were compared between cells incubated in the presence of control exosomes, syncytin-1 or syncytin-2-deprived exosomes, or exosomes solely bearing the uncleaved forms of these HERV env proteins. From our data, we conclude that villous cytotrophoblast exosomes are positive for both env proteins and are rapidly taken up by BeWo cells in a syncytin-1- and syncytin-2-dependent manner and that syncytin-2 is reduced in serum-derived exosomes from women with PE when compared to exosomes from normal pregnant women.


Assuntos
Exossomos/metabolismo , Produtos do Gene env/fisiologia , Pré-Eclâmpsia/sangue , Proteínas da Gravidez/fisiologia , Trofoblastos/metabolismo , Adulto , Sistema ASC de Transporte de Aminoácidos/antagonistas & inibidores , Sistema ASC de Transporte de Aminoácidos/genética , Sistema ASC de Transporte de Aminoácidos/fisiologia , Comunicação Celular , Fusão Celular , Linhagem Celular Tumoral , Coriocarcinoma/patologia , Endocitose , Retrovirus Endógenos/genética , Retrovirus Endógenos/fisiologia , Endossomos/metabolismo , Feminino , Furina/antagonistas & inibidores , Furina/fisiologia , Produtos do Gene env/sangue , Humanos , Microscopia Confocal , Antígenos de Histocompatibilidade Menor , Gravidez , Proteínas da Gravidez/sangue , Proteínas da Gravidez/deficiência , Interferência de RNA , RNA Interferente Pequeno/farmacologia , Simportadores , Proteínas Supressoras de Tumor/antagonistas & inibidores , Proteínas Supressoras de Tumor/genética , Proteínas Supressoras de Tumor/fisiologia , Neoplasias Uterinas/patologia
7.
Cell Signal ; 26(5): 959-67, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24462704

RESUMO

Expression of syncytin-1, or the human endogenous retroviral family W member 1 (HERVWE1) in human placental trophoblasts is regulated by DNA methylation. Increased DNA methylation and decreased expression of syncytin-1 have been observed in preeclamptic placentas. The syncytin-1-mediated fusogenic as well as non-fusogenic activities, e.g., cell cycle promotion, anti-apoptosis, and immune suppression, are implicated in the pathogenic changes in preeclamptic placentas. It is noteworthy that in a close vicinity to syncytin-1 there are two genes, peroxisome biogenesis factor 1 (PEX1) and GATA zinc finger domain containing 1 (GATAD1), as well as multiple CpG islands around these genes. In this study we determined if these adjacent genes might, like syncytin-1, subject to epigenetic regulation in preeclamptic placentas. Data from quantitative real-time PCR and Western blotting indicated that while PEX1 expression remained stable, GATAD1 expression was significantly decreased in the third-trimester placentas associated with preeclampsia than those associated with normal pregnancy. Immunohistochemistry detected high GATAD1 expression in trophoblast linage, and confirmed its reduced levels in preeclamptic placentas. However, COBRA and bisulfate sequencing detected decreased DNA methylation in levels in the 3 [prime] region of GATAD1 gene in preeclamptic placentas. The positive correlation between 3 [prime] methylation and GATAD1 expression was confirmed by treatment of choriocarcinoma JAR cells with DNMT inhibitor. These data pointed to a potential role of GATAD1 for the syncytium deficiency often associated with preeclamptic placentas. The sharp contrast of the methylation alterations for the closely positioned GATAD1 and HERVWE1 may provide a useful model for studying the accurate control of DNA methylation as well as their positive and negative impact on gene expression in placental trophoblasts.


Assuntos
Metilação de DNA , Proteínas do Olho/genética , Proteínas do Olho/metabolismo , Regulação da Expressão Gênica , Placenta/fisiopatologia , Pré-Eclâmpsia/fisiopatologia , ATPases Associadas a Diversas Atividades Celulares , Linhagem Celular Tumoral , Ilhas de CpG , Feminino , Produtos do Gene env/deficiência , Produtos do Gene env/genética , Produtos do Gene env/metabolismo , Humanos , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Placenta/metabolismo , Pré-Eclâmpsia/metabolismo , Pré-Eclâmpsia/patologia , Gravidez , Proteínas da Gravidez/deficiência , Proteínas da Gravidez/genética , Proteínas da Gravidez/metabolismo , Terceiro Trimestre da Gravidez , Análise de Sequência de DNA , Trofoblastos/metabolismo
8.
J Hepatol ; 58(2): 319-28, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23046674

RESUMO

BACKGROUND & AIMS: The placental growth factor (PlGF) is a member of the vascular endothelial growth factor (VEGF) family known to stimulate endothelial cell growth, migration and survival, attract angiocompetent macrophages, and determine the metastatic niche. Unlike VEGF, genetic studies have shown that PlGF is specifically involved in pathologic angiogenesis, thus its inhibition would not affect healthy blood vessels, providing an attractive drug candidate with a good safety profile. METHODS: We assess whether inhibition of PlGF could be used as a potential therapy against hepatocellular carcinoma (HCC), by using PlGF knockout mice and monoclonal anti-PlGF antibodies in a mouse model for HCC. In addition, the effect of PlGF antibodies is compared to that of sorafenib, as well as the combination of both therapies. RESULTS: We have found that both in a transgenic knockout model and in a treatment model, targeting PlGF significantly decreases tumor burden. This was achieved not only by inhibiting neovascularisation, but also by decreasing hepatic macrophage recruitment and by normalising the remaining blood vessels, thereby decreasing hypoxia and reducing the prometastatic potential of HCC. CONCLUSIONS: Considering the favourable safety profile and its pleiotropic effect on vascularisation, metastasis and inflammation, PlGF inhibition could become a valuable therapeutic strategy against HCC.


Assuntos
Carcinoma Hepatocelular/induzido quimicamente , Carcinoma Hepatocelular/fisiopatologia , Dietilnitrosamina/efeitos adversos , Neoplasias Hepáticas/induzido quimicamente , Neoplasias Hepáticas/fisiopatologia , Proteínas da Gravidez/fisiologia , Animais , Anticorpos Monoclonais/imunologia , Anticorpos Monoclonais/farmacologia , Anticorpos Monoclonais/uso terapêutico , Antineoplásicos/farmacologia , Antineoplásicos/uso terapêutico , Carcinoma Hepatocelular/tratamento farmacológico , Modelos Animais de Doenças , Quimioterapia Combinada , Neoplasias Hepáticas/tratamento farmacológico , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Metástase Neoplásica/tratamento farmacológico , Metástase Neoplásica/fisiopatologia , Neovascularização Patológica/tratamento farmacológico , Neovascularização Patológica/fisiopatologia , Niacinamida/análogos & derivados , Niacinamida/farmacologia , Niacinamida/uso terapêutico , Compostos de Fenilureia/farmacologia , Compostos de Fenilureia/uso terapêutico , Fator de Crescimento Placentário , Proteínas da Gravidez/deficiência , Proteínas da Gravidez/imunologia , Sorafenibe , Resultado do Tratamento
9.
Artigo em Inglês | MEDLINE | ID: mdl-22908198

RESUMO

Placental growth factor (PlGF) is a member of the vascular endothelial growth factor (VEGF) family that also comprises VEGF-A (VEGF), VEGF-B, VEGF-C, and VEGF-D. Unlike VEGF, PlGF is dispensable for development and health but has diverse nonredundant roles in tissue ischemia, malignancy, inflammation, and multiple other diseases. Genetic and pharmacological gain-of-function and loss-of-function studies have identified molecular mechanisms of this multitasking cytokine and characterized the therapeutic potential of delivering or blocking PlGF for various disorders.


Assuntos
Citocinas/fisiologia , Proteínas da Gravidez/fisiologia , Animais , Artrite Reumatoide/tratamento farmacológico , Doenças Cardiovasculares/tratamento farmacológico , Doenças Cardiovasculares/etiologia , Colite/etiologia , Citocinas/antagonistas & inibidores , Citocinas/deficiência , Citocinas/genética , Crescimento e Desenvolvimento/fisiologia , Doenças Hematológicas/tratamento farmacológico , Doenças Hematológicas/etiologia , Humanos , Hipersensibilidade Tardia/tratamento farmacológico , Cirrose Hepática/tratamento farmacológico , Camundongos , Neoplasias/tratamento farmacológico , Neoplasias/etiologia , Obesidade/tratamento farmacológico , Fator de Crescimento Placentário , Proteínas da Gravidez/antagonistas & inibidores , Proteínas da Gravidez/deficiência , Proteínas da Gravidez/genética , Enfisema Pulmonar/tratamento farmacológico , Sepse/etiologia , Transdução de Sinais/fisiologia , Receptor 1 de Fatores de Crescimento do Endotélio Vascular/fisiologia , Cicatrização/fisiologia
10.
Placenta ; 33(9): 663-71, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22695103

RESUMO

During their replication, infectious retroviruses insert a reverse-transcribed cDNA copy of their genome, a "provirus", into the genome of their host. If the infected cell belongs to the germline, the integrated provirus can become "fixed" within the host genome as an endogenous retrovirus and be transmitted vertically to the progeny in a Mendelian fashion. Based on the numerous proviral sequences that are recovered within the genomic DNA of vertebrates--up to ten percent in the case of mammals--such events must have occurred repeatedly during the course of millions of years of evolution. Although most of the ancient proviral sequences have been disrupted, a few "endogenized" retroviral genes are conserved and still encode functional proteins. In this review, we focus on the recent discovery of genes derived from the envelope glycoprotein-encoding (env) genes of endogenous retroviruses that have been domesticated by mammals to carry out an essential function in placental development. They were called syncytins based on the membrane fusogenic capacity that they have kept from their parental env gene and which contributes to the formation of the placental fused cell layer called the syncytiotrophoblast, at the materno-fetal interface. Remarkably, the capture of syncytin or syncytin-like genes, sometimes as pairs, was found to have occurred independently from different endogenous retroviruses in diverse mammalian lineages such as primates--including humans--, muroids, leporids, carnivores, caviids, and ovis, between around 10 and 85 million years ago. Knocking out one or both mouse syncytin-A and -B genes provided evidence that they indeed play a critical role in placentation. We discuss the possibility that the immunosuppressive domain embedded within retroviral envelope glycoproteins and conserved in syncytin proteins, may be involved in the tolerance of the fetus by the maternal immune system. Finally, we speculate that the capture of a founding syncytin-like gene could have been instrumental in the dramatic transition from egg-laying to placental mammals.


Assuntos
Produtos do Gene env/genética , Produtos do Gene env/fisiologia , Placentação/fisiologia , Proteínas da Gravidez/genética , Proteínas da Gravidez/fisiologia , Retroviridae/genética , Integração Viral/genética , Animais , Sequência de Bases , Evolução Biológica , Sequência Conservada , Retrovirus Endógenos/genética , Feminino , Humanos , Tolerância Imunológica , Camundongos , Camundongos Knockout , Placentação/genética , Gravidez , Proteínas da Gravidez/deficiência , Proteínas do Envelope Viral/genética , Proteínas do Envelope Viral/fisiologia
11.
J Immunol ; 185(11): 7085-96, 2010 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-20974989

RESUMO

Uterine dendritic cells (DCs) are critical for activating the T cell response mediating maternal immune tolerance of the semiallogeneic fetus. GM-CSF (CSF2), a known regulator of DCs, is synthesized by uterine epithelial cells during induction of tolerance in early pregnancy. To investigate the role of GM-CSF in regulating uterine DCs and macrophages, Csf2-null mutant and wild-type mice were evaluated at estrus, and in the periconceptual and peri-implantation periods. Immunohistochemistry showed no effect of GM-CSF deficiency on numbers of uterine CD11c(+) cells and F4/80(+) macrophages at estrus or on days 0.5 and 3.5 postcoitum, but MHC class II(+) and class A scavenger receptor(+) cells were fewer. Flow cytometry revealed reduced CD80 and CD86 expression by uterine CD11c(+) cells and reduced MHC class II in both CD11c(+) and F4/80(+) cells from GM-CSF-deficient mice. CD80 and CD86 were induced in Csf2(-/-) uterine CD11c(+) cells by culture with GM-CSF. Substantially reduced ability to activate both CD4(+) and CD8(+) T cells in vivo was evident after delivery of OVA Ag by mating with Act-mOVA males or transcervical administration of OVA peptides. This study shows that GM-CSF regulates the efficiency with which uterine DCs and macrophages activate T cells, and it is essential for optimal MHC class II- and class I-mediated indirect presentation of reproductive Ags. Insufficient GM-CSF may impair generation of T cell-mediated immune tolerance at the outset of pregnancy and may contribute to the altered DC profile and dysregulated T cell tolerance evident in infertility, miscarriage, and preeclampsia.


Assuntos
Células Dendríticas/imunologia , Células Dendríticas/metabolismo , Fator Estimulador de Colônias de Granulócitos e Macrófagos/fisiologia , Ativação Linfocitária/imunologia , Proteínas da Gravidez/fisiologia , Subpopulações de Linfócitos T/imunologia , Útero/imunologia , Útero/metabolismo , Sequência de Aminoácidos , Animais , Células Apresentadoras de Antígenos/imunologia , Células Apresentadoras de Antígenos/metabolismo , Endométrio/embriologia , Endométrio/imunologia , Endométrio/metabolismo , Epitopos de Linfócito T/imunologia , Feminino , Fator Estimulador de Colônias de Granulócitos e Macrófagos/deficiência , Fator Estimulador de Colônias de Granulócitos e Macrófagos/genética , Antígenos de Histocompatibilidade Classe I/metabolismo , Antígenos de Histocompatibilidade Classe II/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Dados de Sequência Molecular , Especificidade de Órgãos/imunologia , Gravidez , Proteínas da Gravidez/deficiência , Proteínas da Gravidez/genética , Subpopulações de Linfócitos T/metabolismo , Útero/embriologia
12.
J Immunol ; 184(6): 3202-12, 2010 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-20147628

RESUMO

Proinflammatory CC chemokines are thought to drive recruitment of maternal leukocytes into gestational tissues and regulate extravillous trophoblast migration. The atypical chemokine receptor D6 binds many of these chemokines and is highly expressed by the human placenta. D6 is thought to act as a chemokine scavenger because, when ectopically expressed in cell lines in vitro, it efficiently internalizes proinflammatory CC chemokines and targets them for destruction in the absence of detectable chemokine-induced signaling. Moreover, D6 suppresses inflammation in many mouse tissues, and notably, D6-deficient fetuses in D6-deficient female mice show increased susceptibility to inflammation-driven resorption. In this paper, we report strong anti-D6 immunoreactivity, with specific intracellular distribution patterns, in trophoblast-derived cells in human placenta, decidua, and gestational membranes throughout pregnancy and in trophoblast disease states of hydatidiform mole and choriocarcinoma. We show, for the first time, that endogenous D6 in a human choriocarcinoma-derived cell line can mediate progressive chemokine scavenging and that the D6 ligand CCL2 can specifically associate with human syncytiotrophoblasts in term placenta in situ. Moreover, despite strong chemokine production by gestational tissues, levels of D6-binding chemokines in maternal plasma decrease during pregnancy, even in women with pre-eclampsia, a disease associated with increased maternal inflammation. In mice, D6 is not required for syngeneic or semiallogeneic fetal survival in unchallenged mice, but interestingly, it does suppress fetal resorption after embryo transfer into fully allogeneic recipients. These data support the view that trophoblast D6 scavenges maternal chemokines at the fetomaternal interface and that, in some circumstances, this can help to ensure fetal survival.


Assuntos
Transferência Embrionária , Embrião de Mamíferos/imunologia , Sobrevivência de Enxerto/imunologia , Proteínas da Gravidez/genética , Receptores CCR10/genética , Animais , Linhagem Celular Tumoral , Quimiocina CCL2/sangue , Regulação para Baixo/genética , Regulação para Baixo/imunologia , Transferência Embrionária/efeitos adversos , Transferência Embrionária/mortalidade , Embrião de Mamíferos/citologia , Embrião de Mamíferos/metabolismo , Feminino , Sobrevivência de Enxerto/genética , Humanos , Mediadores da Inflamação/sangue , Mediadores da Inflamação/metabolismo , Masculino , Troca Materno-Fetal/genética , Troca Materno-Fetal/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos DBA , Pré-Eclâmpsia/genética , Pré-Eclâmpsia/imunologia , Pré-Eclâmpsia/patologia , Gravidez , Resultado da Gravidez/genética , Proteínas da Gravidez/biossíntese , Proteínas da Gravidez/sangue , Proteínas da Gravidez/deficiência , Ligação Proteica/genética , Ligação Proteica/imunologia , Receptores CCR10/biossíntese , Receptores CCR10/sangue , Receptores CCR10/deficiência , Transplante Homólogo/mortalidade , Trofoblastos/citologia , Trofoblastos/imunologia , Trofoblastos/metabolismo , Receptor D6 de Quimiocina
13.
J Biol Chem ; 285(3): 2140-51, 2010 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-19901022

RESUMO

We identified a sequence homologous to the Bcl-2 homology 3 (BH3) domain of Bcl-2 proteins in SOUL. Tissues expressed the protein to different extents. It was predominantly located in the cytoplasm, although a fraction of SOUL was associated with the mitochondria that increased upon oxidative stress. Recombinant SOUL protein facilitated mitochondrial permeability transition and collapse of mitochondrial membrane potential (MMP) and facilitated the release of proapoptotic mitochondrial intermembrane proteins (PMIP) at low calcium and phosphate concentrations in a cyclosporine A-dependent manner in vitro in isolated mitochondria. Suppression of endogenous SOUL by diced small interfering RNA in HeLa cells increased their viability in oxidative stress. Overexpression of SOUL in NIH3T3 cells promoted hydrogen peroxide-induced cell death and stimulated the release of PMIP but did not enhance caspase-3 activation. Despite the release of PMIP, SOUL facilitated predominantly necrotic cell death, as revealed by annexin V and propidium iodide staining. This necrotic death could be the result of SOUL-facilitated collapse of MMP demonstrated by JC-1 fluorescence. Deletion of the putative BH3 domain sequence prevented all of these effects of SOUL. Suppression of cyclophilin D prevented these effects too, indicating that SOUL facilitated mitochondrial permeability transition in vivo. Overexpression of Bcl-2 and Bcl-x(L), which can counteract the mitochondria-permeabilizing effect of BH3 domain proteins, also prevented SOUL-facilitated collapse of MMP and cell death. These data indicate that SOUL can be a novel member of the BH3 domain-only proteins that cannot induce cell death alone but can facilitate both outer and inner mitochondrial membrane permeabilization and predominantly necrotic cell death in oxidative stress.


Assuntos
Permeabilidade da Membrana Celular , Hemeproteínas/química , Hemeproteínas/metabolismo , Membranas Mitocondriais/metabolismo , Estresse Oxidativo , Proteínas da Gravidez/química , Proteínas da Gravidez/metabolismo , Proteínas Proto-Oncogênicas c-bcl-2/química , Homologia de Sequência de Aminoácidos , Sequência de Aminoácidos , Animais , Apoptose/efeitos dos fármacos , Apoptose/genética , Bovinos , Permeabilidade da Membrana Celular/efeitos dos fármacos , Sobrevivência Celular , Peptidil-Prolil Isomerase F , Ciclofilinas/farmacologia , Regulação da Expressão Gênica , Células HeLa , Proteínas Ligantes de Grupo Heme , Hemeproteínas/deficiência , Hemeproteínas/genética , Humanos , Peróxido de Hidrogênio/farmacologia , Potencial da Membrana Mitocondrial/efeitos dos fármacos , Camundongos , Membranas Mitocondriais/efeitos dos fármacos , Proteínas Mitocondriais/metabolismo , Dados de Sequência Molecular , Células NIH 3T3 , Estresse Oxidativo/efeitos dos fármacos , Proteínas da Gravidez/deficiência , Proteínas da Gravidez/genética , Estrutura Terciária de Proteína , RNA Interferente Pequeno/genética , Ratos , Deleção de Sequência
14.
Respir Res ; 10: 115, 2009 Nov 23.
Artigo em Inglês | MEDLINE | ID: mdl-19930612

RESUMO

BACKGROUND: Although both animal and human studies suggested the association between placenta growth factor (PlGF) and chronic obstructive pulmonary disease (COPD), especially lung emphysema, the role of PlGF in the pathogenesis of emphysema remains to be clarified. This study hypothesizes that blocking PlGF prevents the development of emphysema. METHODS: Pulmonary emphysema was induced in PlGF knock-out (KO) and wild type (WT) mice by intra-tracheal instillation of porcine pancreatic elastase (PPE). A group of KO mice was then treated with exogenous PlGF and WT mice with neutralizing anti-VEGFR1 antibody. Tumor necrosis factor alpha (TNF-alpha), matrix metalloproteinase-9 (MMP-9), and VEGF were quantified. Apoptosis measurement and immunohistochemical staining for VEGF R1 and R2 were performed in emphysematous lung tissues. RESULTS: After 4 weeks of PPE instillation, lung airspaces enlarged more significantly in WT than in KO mice. The levels of TNF-alpha and MMP-9, but not VEGF, increased in the lungs of WT compared with those of KO mice. There was also increased in apoptosis of alveolar septal cells in WT mice. Instillation of exogenous PlGF in KO mice restored the emphysematous changes. The expression of both VEGF R1 and R2 decreased in the emphysematous lungs. CONCLUSION: In this animal model, pulmonary emphysema is prevented by depleting PlGF. When exogenous PlGF is administered to PlGF KO mice, emphysema re-develops, implying that PlGF contributes to the pathogenesis of emphysema.


Assuntos
Pulmão/metabolismo , Proteínas da Gravidez/deficiência , Enfisema Pulmonar/prevenção & controle , Animais , Apoptose , Modelos Animais de Doenças , Pulmão/patologia , Metaloproteinase 9 da Matriz/metabolismo , Camundongos , Camundongos Knockout , Elastase Pancreática , Fator de Crescimento Placentário , Proteínas da Gravidez/genética , Enfisema Pulmonar/induzido quimicamente , Enfisema Pulmonar/metabolismo , Enfisema Pulmonar/patologia , Fator de Necrose Tumoral alfa/metabolismo , Fator A de Crescimento do Endotélio Vascular/metabolismo , Receptor 1 de Fatores de Crescimento do Endotélio Vascular/metabolismo , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo
15.
Gastroenterology ; 137(6): 2112-24.e1-6, 2009 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19751735

RESUMO

BACKGROUND & AIMS: Portal hypertension is responsible for the major complications associated with cirrhosis. Angiogenesis has been associated with the pathophysiology of portal hypertension. We investigated the role of placental growth factor (PlGF) and tested the effects of monoclonal antibodies against PlGF (alphaPlGF) in a mouse model of portal hypertension. METHODS: Using a mouse model of prehepatic portal hypertension, we measured PlGF levels in the mesenteric tissue at different time points. We used knockout mice and alphaPlGF to determine the role of PlGF in the splanchnic hyperdynamic system and portosystemic collateral formation, examining its effects before and after portal hypertension was induced. RESULTS: PlGF was significantly up-regulated in the mesenteric tissue of mice with portal hypertension. Compared with wild-type animals, the vascular density in the mesentery was reduced in PlGF knockout hypertensive mice, preventing collateral formation and attenuation of mesenteric artery flow without affecting portal pressure. In the prevention study, alphaPlGF showed similar findings as in the knockout study. In mice with portal hypertension, administration of alphaPlGF resulted in a 32% decrease in portal pressure, compared with mice given immunoglobulin G(1) (control). CONCLUSIONS: Pathologic angiogenesis in the mesenteric tissues of mice with portal hypertension is mediated by PlGF. Blocking PlGF could be an effective strategy for reducing collateral formation and lowering portal pressure; further research into the effects in cirrhosis is warranted.


Assuntos
Circulação Colateral , Hipertensão Portal/metabolismo , Mesentério/irrigação sanguínea , Mesentério/metabolismo , Neovascularização Patológica/metabolismo , Proteínas da Gravidez/metabolismo , Circulação Esplâncnica , Proteínas Angiogênicas/metabolismo , Animais , Anticorpos Monoclonais/farmacologia , Anti-Hipertensivos/farmacologia , Circulação Colateral/efeitos dos fármacos , Modelos Animais de Doenças , Hipertensão Portal/tratamento farmacológico , Hipertensão Portal/fisiopatologia , Cinética , Masculino , Camundongos , Camundongos Knockout , Microcirculação , Neovascularização Patológica/fisiopatologia , Neovascularização Patológica/prevenção & controle , Fator de Crescimento Placentário , Pressão na Veia Porta , Proteínas da Gravidez/deficiência , Proteínas da Gravidez/genética , Proteínas da Gravidez/imunologia , Transdução de Sinais , Circulação Esplâncnica/efeitos dos fármacos , Regulação para Cima
17.
Proc Natl Acad Sci U S A ; 106(29): 12127-32, 2009 Jul 21.
Artigo em Inglês | MEDLINE | ID: mdl-19564597

RESUMO

In most mammalian species, a key process of placenta development is the fusion of trophoblast cells into a highly specialized, multinucleated syncytiotrophoblast layer, through which most of the maternofetal exchanges take place. Little is known about this process, despite the recent identification of 2 pairs of envelope genes of retroviral origin, independently acquired by the human (syncytin-1 and syncytin-2) and mouse (syncytin-A and syncytin-B) genomes, specifically expressed in the placenta, and with in vitro cell-cell fusion activity. By generating knockout mice, we show here that homozygous syncytin-A null mouse embryos die in utero between 11.5 and 13.5 days of gestation. Refined cellular and subcellular analyses of the syncytin-A-deficient placentae disclose specific disruption of the architecture of the syncytiotrophoblast-containing labyrinth, with the trophoblast cells failing to fuse into an interhemal syncytial layer. Lack of syncytin-A-mediated trophoblast cell fusion is associated with cell overexpansion at the expense of fetal blood vessel spaces and with apoptosis, adding to the observed maternofetal interface structural defects to provoke decreased vascularization, inhibition of placental transport, and fetal growth retardation, ultimately resulting in death of the embryo. These results demonstrate that syncytin-A is essential for trophoblast cell differentiation and syncytiotrophoblast morphogenesis during placenta development, and they provide evidence that genes captured from ancestral retroviruses have been pivotal in the acquisition of new, important functions in mammalian evolution.


Assuntos
Retrovirus Endógenos/genética , Placentação/fisiologia , Proteínas da Gravidez/deficiência , Proteínas do Envelope Viral/genética , Animais , Cruzamentos Genéticos , Perda do Embrião/metabolismo , Perda do Embrião/patologia , Embrião de Mamíferos/anormalidades , Embrião de Mamíferos/patologia , Membranas Extraembrionárias/anormalidades , Membranas Extraembrionárias/patologia , Feminino , Marcação de Genes , Genótipo , Masculino , Camundongos , Camundongos Knockout , Placenta/anormalidades , Placenta/patologia , Placenta/ultraestrutura , Gravidez , Proteínas da Gravidez/metabolismo , Trofoblastos/patologia
18.
Mol Reprod Dev ; 75(1): 175-83, 2008 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-17546632

RESUMO

Preeclampsia (PE), Hemolysis Elevated Liver Enzymes and Low Platelets (HELLP)-syndrome, and intrauterine growth restriction (IUGR) are associated with abnormal placentation. In early pregnancy, placental cytotrophoblasts fuse and form multinuclear syncytiotrophoblasts. The envelope gene of the human endogenous retrovirus-W, Syncytin, is a key factor for mediating cell-cell fusion of cytotrophoblasts. This study investigated clinical parameters of PE and HELLP-associated IUGR and analyzed the cell-cell fusion index and beta-human chorionic gonadotropin (beta-hCG) secretion of cytotrophoblasts isolated and cultured from placentas of these patients. In addition, we performed absolute quantitation of Syncytin and determined the apoptosis rate in both cultured cytotrophoblasts and placental tissues. Cultured cytotrophoblasts from PE and HELLP-associated IUGR correlated with a pronounced lower cell-cell fusion index, 1.8- and 3.6-fold; less nuclei per syncytiotrophoblast, 1.4- and 2.0-fold; a significantly decreased beta-hCG secretion, 4.3- and 17.2-fold and a reduction of Syncytin gene expression, 8.1 (P = 0.019) and 222.7-fold (P = 0.011) compared with controls, respectively. In contrast, a significantly 2.3-fold higher apoptosis rate was observed in cultured PE/IUGR cytotrophoblasts (P = 0.043). Importantly, Syncytin gene expression in primary placental tissues of PE/IUGR was 5.4-fold lower (P = 0.047) and in HELLP/IUGR 10.6-fold lower (P = 0.019) along with a 1.8- and 1.9-fold significant increase in the apoptosis rate compared with controls, respectively. Low Syncytin expression in both cultured cytotrophoblasts and primary tissues from pathological placentas supports an intrinsic placenta-specific deregulation of cell-cell fusion in the formation of syncytiotrophoblasts leading to increased apoptosis. These processes could contribute to the development and severity of PE and HELLP-associated IUGR.


Assuntos
Retardo do Crescimento Fetal/etiologia , Produtos do Gene env/deficiência , Síndrome HELLP/patologia , Placenta/patologia , Pré-Eclâmpsia/patologia , Proteínas da Gravidez/deficiência , Trofoblastos/patologia , Adulto , Apoptose , Fusão Celular , Células Cultivadas , Gonadotropina Coriônica Humana Subunidade beta/genética , Gonadotropina Coriônica Humana Subunidade beta/metabolismo , Feminino , Retardo do Crescimento Fetal/metabolismo , Expressão Gênica , Produtos do Gene env/genética , Síndrome HELLP/metabolismo , Humanos , Placenta/metabolismo , Pré-Eclâmpsia/metabolismo , Gravidez , Proteínas da Gravidez/genética , Trofoblastos/metabolismo
19.
Proc Natl Acad Sci U S A ; 103(40): 14947-52, 2006 Oct 03.
Artigo em Inglês | MEDLINE | ID: mdl-17003132

RESUMO

The presence of the envelope glycoprotein Env in HIV-1 virions is essential for infectivity. To date, the molecular mechanism by which Env is packaged into virions has been largely unknown. Here, we show that TIP47 (tail-interacting protein of 47 kDa), which has been shown to interact with Env, also binds the MA (matrix) domain of HIV-1 Gag protein and that these three proteins form a ternary complex. Mutations in Gag that abrogate interaction with TIP47 inhibit Env incorporation and virion infectivity as well as colocalization between Gag and Env. We also show that TIP47 silencing impairs Env incorporation and infectivity and abolishes coimmunoprecipitation of Gag with Env. In contrast, overexpression of TIP47 increases Env packaging. Last, we demonstrate that TIP47 can interact simultaneously with Env and Gag. Taken together, our results show that TIP47 is a cellular cofactor that plays an essential role in Env incorporation, allowing the encounter and the physical association between HIV-1 Gag and Env proteins during the viral assembly process.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Produtos do Gene env/metabolismo , Produtos do Gene gag/metabolismo , HIV-1/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Proteínas da Gravidez/metabolismo , Vírion/metabolismo , Sequência de Aminoácidos , Aminoácidos , Proteínas de Ligação a DNA/deficiência , Expressão Gênica , Proteína gp41 do Envelope de HIV/metabolismo , HIV-1/fisiologia , Células HeLa , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/deficiência , Células Jurkat , Dados de Sequência Molecular , Mutação/genética , Perilipina-3 , Proteínas da Gravidez/deficiência , Ligação Proteica , Estrutura Terciária de Proteína , Proteínas de Transporte Vesicular , Proteínas da Matriz Viral/química
20.
Diabetes ; 55(10): 2698-704, 2006 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-17003333

RESUMO

Placental growth factor (PlGF)-deficient (PlGF-/-) and wild-type mice were kept on a standard-fat or high-fat diet for 15 weeks. With the standard-fat diet, the body weights of PlGF-/- and wild-type mice were comparable, whereas the combined weight of subcutaneous and gonadal adipose tissues was lower in PlGF-/- mice (P = 0.02). With the high-fat diet, PlGF-/- mice had a lower body weight (P < 0.05) and less total subcutaneous plus gonadal adipose tissue (P < 0.0001). Blood vessel size was lower in gonadal adipose tissue of PlGF-/- mice with both the standard-fat and high-fat diet (P < 0.05). Blood vessel density, normalized to adipocyte number, was significantly lower in subcutaneous adipose tissue of PlGF-/- mice fed the high-fat diet (P < 0.01). De novo adipose tissue development in nude mice injected with 3T3-F442A preadipocytes was reduced (P < 0.005) by administration of a PlGF-neutralizing antibody. Bone marrow transplantation from wild-type or PlGF-/- mice to wild-type or PlGF-/- recipient mice revealed significantly lower blood vessel density in PlGF-/- recipient mice without an effect on adipose tissue growth. Thus, in murine models of diet-induced obesity, inactivation of PlGF impairs adipose tissue development, at least in part as a result of reduced angiogenesis.


Assuntos
Tecido Adiposo/crescimento & desenvolvimento , Proteínas da Gravidez/deficiência , Adipócitos/citologia , Tecido Adiposo/irrigação sanguínea , Animais , Anticorpos Monoclonais/farmacologia , Transplante de Medula Óssea/fisiologia , Gorduras na Dieta/administração & dosagem , Masculino , Camundongos , Camundongos Nus , Fator de Crescimento Placentário , Proteínas da Gravidez/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA