Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 36
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Development ; 147(21)2020 09 02.
Artigo em Inglês | MEDLINE | ID: mdl-32878913

RESUMO

Temple and Kagami-Ogata syndromes are genomic imprinting diseases caused by maternal and paternal duplication of human chromosome 14, respectively. They exhibit different postnatal muscle-related symptoms as well as prenatal placental problems. Using the mouse models for these syndromes, it has been demonstrated that retrotransposon gag like 1 [Rtl1, also known as paternally expressed 11 (Peg11)] located in the mouse orthologous imprinted region is responsible for the prenatal placental problems because it is an essential placental gene for maintenance of fetal capillary network during gestation. However, the causative imprinted gene for the postnatal muscle-related symptoms remains unknown. Here, we demonstrate that Rtl1 also plays an important role in fetal/neonatal skeletal muscle development: its deletion and overproduction in mice lead to neonatal lethality associated with severe but distinct skeletal muscle defects, similar to those of Temple and Kagami-Ogata syndromes, respectively. Thus, it is strongly suggested that RTL1 is the major gene responsible for the muscle defects in addition to the placental defects in these two genomic imprinting diseases. This is the first example of an LTR retrotransposon-derived gene specific to eutherians contributing to eutherian skeletal muscle development.


Assuntos
Anormalidades Múltiplas/metabolismo , Anormalidades Múltiplas/patologia , Músculos/anormalidades , Proteínas da Gravidez/deficiência , Animais , Animais Recém-Nascidos , Diferenciação Celular , Proliferação de Células , Desmina/metabolismo , Feminino , Feto/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Humanos , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Modelos Genéticos , Fibras Musculares Esqueléticas/metabolismo , Fibras Musculares Esqueléticas/patologia , Músculos/embriologia , Músculos/patologia , Mutação/genética , Proteínas da Gravidez/genética , Proteínas da Gravidez/metabolismo , Células Satélites de Músculo Esquelético/metabolismo , Síndrome , Fatores de Tempo
2.
Mol Reprod Dev ; 86(6): 696-713, 2019 06.
Artigo em Inglês | MEDLINE | ID: mdl-31012985

RESUMO

Prolactin family 7, subfamily d, member 1 (Prl7d1), a member of the expanding prolactin family, is mainly expressed in the placental junctional zone (including trophoblast giant cells and spongiotrophoblast cells) with peak expression observed at 12 days postcoitum (dpc) in mice. Previous studies have shown that PRL7D1 is a key mediator of angiogenesis in vitro; however, its physiological roles in placental development in vivo have not been characterized. To address this issue, we deleted Prl7d1 in mice and demonstrated that its absence results in reduced litter size and fertility. Histologically, Prl7d1 mutants exhibited striking placental abnormalities at 12.5 dpc, including a reduction in the proportion of labyrinth layers and a significant increase in decidual natural killer cells, glycogen trophoblasts, and trophoblast giant cells in the junctional zone. Moreover, placentas from Prl7d1-null mice displayed a thickened decidual spiral artery. Notably, these negative effects were more pronounced in male fetuses. Further RNA-sequencing analysis showed that Prl7d1 deletion results in significant differences in the placental transcriptome profile between the two sexes of fetuses. Together, this study demonstrates that Prl7d1 possesses antiangiogenic properties in deciduas and inhibits the development of junctional zone, which potentially alters the functional capacity of the placenta to support optimal fetal growth. Moreover, of note, the role of Prl7d1 in the placenta is regulated in a fetal sex-specific manner.


Assuntos
Desenvolvimento Fetal , Feto/embriologia , Deleção de Genes , Doenças Placentárias/metabolismo , Placenta/metabolismo , Proteínas da Gravidez/deficiência , Caracteres Sexuais , Animais , Feminino , Feto/patologia , Masculino , Camundongos , Camundongos Knockout , Placenta/patologia , Doenças Placentárias/patologia , Gravidez , Proteínas da Gravidez/metabolismo
3.
J Inherit Metab Dis ; 42(5): 809-817, 2019 09.
Artigo em Inglês | MEDLINE | ID: mdl-31177572

RESUMO

The first step in branched-chain amino acid (BCAA) catabolism is catalyzed by the two BCAA transferase isoenzymes, cytoplasmic branched-chain amino acid transferase (BCAT) 1, and mitochondrial BCAT2. Defects in the second step of BCAA catabolism cause maple syrup urine disease (MSUD), a condition which has been far more extensively investigated. Here, we studied the consequences of BCAT2 deficiency, an ultra-rare condition in humans. We present genetic, clinical, and functional data in five individuals from four different families with homozygous or compound heterozygous BCAT2 mutations which were all detected following abnormal biochemical profile results or familial mutation segregation studies. We demonstrate that BCAT2 deficiency has a recognizable biochemical profile with raised plasma BCAAs and, in contrast with MSUD, low-normal branched-chain keto acids (BCKAs) with undetectable l-allo-isoleucine. Interestingly, unlike in MSUD, none of the individuals with BCAT2 deficiency developed acute encephalopathy even with exceptionally high BCAA levels. We observed wide-ranging clinical phenotypes in individuals with BCAT2 deficiency. While one adult was apparently asymptomatic, three individuals had presented with developmental delay and autistic features. We show that the biochemical characteristics of BCAT2 deficiency may be amenable to protein-restricted diet and that early treatment may improve outcome in affected individuals. BCAT2 deficiency is an inborn error of BCAA catabolism. At present, it is unclear whether developmental delay and autism are parts of the variable phenotypic spectrum of this condition or coincidental. Further studies will be required to explore this.


Assuntos
Erros Inatos do Metabolismo dos Aminoácidos/diagnóstico , Erros Inatos do Metabolismo dos Aminoácidos/genética , Aminoácidos de Cadeia Ramificada/sangue , Encéfalo/patologia , Mitocôndrias/patologia , Proteínas da Gravidez/deficiência , Transaminases/deficiência , Adolescente , Adulto , Encéfalo/diagnóstico por imagem , Criança , Pré-Escolar , Feminino , Homozigoto , Humanos , Imageamento por Ressonância Magnética , Masculino , Antígenos de Histocompatibilidade Menor/genética , Mutação , Fenótipo , Proteínas da Gravidez/genética , Transaminases/genética
4.
Genes Cells ; 22(2): 174-188, 2017 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-28111885

RESUMO

Paternally expressed 11/Retrotransposon-like 1 (Peg11/Rtl1) knockout (KO) mice show mid- to late fetal lethality or late fetal growth retardation associated with frequent neonatal lethality. The lethal phenotype is largely dependent on genetic background and becomes more severe with each succeeding generation in the course of backcross experiments to C57BL/6 (B6). We previously suggested that these lethal and growth phenotypes in the fetal stages were due to severe defects in placental fetal capillaries in the labyrinth layer. In this study, we re-examined KO fetuses and placentas and confirmed that the severe clogging of fetal capillaries was associated with KO fetuses showing mid-fetal lethality with internal bleeding. Importantly, the basal region of the fetal capillary network was specifically damaged, also leading to poor expansion of the labyrinth layer and placental size reduction in the later stage. An apparent down-regulation of transmembrane protein 100 (Tmem100), mesenchyme homeobox 2 (Meox2) and lymphatic vessel endothelial hyaluronan receptor 1 (Lyve1) expression were observed in earlier stage placentas even before apparent size reduction became, suggesting that these genes are involved in the maintenance of fetal capillaries associated with Peg11/Rtl1 during development.


Assuntos
Placenta/irrigação sanguínea , Proteínas da Gravidez/genética , Proteínas da Gravidez/metabolismo , Animais , Capilares/metabolismo , Capilares/patologia , Feminino , Morte Fetal , Retardo do Crescimento Fetal/etiologia , Masculino , Camundongos , Camundongos Knockout , Placenta/anatomia & histologia , Placenta/metabolismo , Gravidez , Proteínas da Gravidez/deficiência
5.
Biol Reprod ; 94(1): 6, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26586843

RESUMO

The X-linked Plac1 gene is maternally expressed in trophoblast cells during placentation, and its disruption causes placental hyperplasia and intrauterine growth restriction. In contrast, Plac1 is also reported to be one of the upregulated genes in the hyperplastic placenta generated by nuclear transfer. However, the effect of overexpressed Plac1 on placental formation and function remained unaddressed. We complemented the Plac1 knockout placental dysfunction by lentiviral vector-mediated, placenta-specific Plac1 transgene expression. Whereas fetal development and the morphology of maternal blood sinuses in the labyrinth zone improved, placental hyperplasia remained, with an expanded the junctional zone that migrated and encroached into the labyrinth zone. Further experiments revealed that wild-type placenta with transgenically expressed Plac1 resulted in placental hyperplasia without the encroaching of the junctional zone. Our findings suggest that Plac1 is involved in trophoblast cell proliferation, differentiation, and migration. Its proper expression is required for normal placentation and fetal development.


Assuntos
Viabilidade Fetal/genética , Lentivirus/genética , Placenta/patologia , Proteínas da Gravidez/deficiência , Proteínas da Gravidez/genética , Animais , Blastocisto/metabolismo , Proliferação de Células , Feminino , Retardo do Crescimento Fetal/genética , Retardo do Crescimento Fetal/patologia , Regulação da Expressão Gênica no Desenvolvimento , Teste de Complementação Genética , Vetores Genéticos , Hiperplasia , Camundongos , Camundongos Knockout , Técnicas de Transferência Nuclear , Gravidez , Transgenes/genética , Trofoblastos
6.
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
7.
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
8.
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
9.
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
10.
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
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.
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
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.
Glia ; 59(3): 379-96, 2011 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21264946

RESUMO

Wallerian degeneration (WD) is an inflammatory process of nerve degeneration, which occurs more rapidly in the peripheral nervous system compared with the central nervous system, resulting, respectively in successful and aborted axon regeneration. In the peripheral nervous system, Schwann cells (SCs) and macrophages, under the control of a network of cytokines and chemokines, represent the main cell types involved in this process. Within this network, the role of placental growth factor (PlGF) remains totally unknown. However, properties like monocyte activation/attraction, ability to increase expression of pro-inflammatory molecules, as well as neuroprotective effects, make it a candidate likely implicated in this process. Also, nothing is described about the expression and localization of this molecule in the peripheral nervous system. To address these original questions, we decided to study PlGF expression under physiological and degenerative conditions and to explore its role in WD, using a model of sciatic nerve transection in wild-type and Pgf(-/-) mice. Our data show dynamic changes of PlGF expression, from periaxonal in normal nerve to SCs 24h postinjury, in parallel with a p65/NF-κB recruitment on Pgf promoter. After injury, SC proliferation is reduced by 30% in absence of PlGF. Macrophage invasion is significantly delayed in Pgf(-/-) mice compared with wild-type mice, which results in worse functional recovery. MCP-1 and proMMP-9 exhibit a 3-fold reduction of their relative expressions in Pgf(-/-) injured nerves, as demonstrated by cytokine array. In conclusion, this work originally describes PlGF as a novel member of the cytokine network of WD.


Assuntos
Fibras Nervosas Mielinizadas/fisiologia , Proteínas da Gravidez/fisiologia , Células de Schwann/metabolismo , Neuropatia Ciática/metabolismo , Degeneração Walleriana/metabolismo , Animais , Células Cultivadas , Citocinas/deficiência , Citocinas/fisiologia , Modelos Animais de Doenças , Feminino , Camundongos , Camundongos Knockout , Simulação de Dinâmica Molecular , Fibras Nervosas Mielinizadas/patologia , Fator de Crescimento Placentário , Proteínas da Gravidez/deficiência , Células de Schwann/patologia , Neuropatia Ciática/patologia , Degeneração Walleriana/patologia
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
16.
J Neurosci ; 28(15): 3877-86, 2008 Apr 09.
Artigo em Inglês | MEDLINE | ID: mdl-18400886

RESUMO

Prion protein (PrP) is a normal component of neurons, which confers susceptibility to prion diseases. Despite its evolutionary conservation, its normal function remains controversial. PrP-deficient (Prnp(0/0)) mice have weaker afterhyperpolarizations (AHPs) in cerebellar and hippocampal neurons. Here we show that the AHP impairment in hippocampal CA1 pyramidal cells is selective for the slow AHP, and is not caused by an impairment of either voltage-gated Ca(2+) channels or Ca(2+)-activated K(+) channels. Instead, Prnp(0/0) neurons have twofold to threefold stronger Ca(2+) buffering and double the Ca(2+) extrusion rate. In Prnp(0/0) neurons thapsigargin abolished the stronger Ca(2+) buffering and extrusion, and thapsigargin or cyclopiazonic acid abolished the weakening of the slow AHPs. These data implicate sarcoplasmic/endoplasmic reticulum calcium ATPase in the enhanced Ca(2+) buffering, and extrusion into the endoplasmic reticulum, which contains substantial amounts of PrP in wild-type mice. Altered Ca(2+) homeostasis can explain several phenotypes identified in Prnp(0/0) mice.


Assuntos
Cálcio/metabolismo , Hipocampo/fisiologia , Proteínas da Gravidez/deficiência , Células Piramidais , Potenciais de Ação , Animais , Soluções Tampão , Sinalização do Cálcio , Eletrofisiologia , Inibidores Enzimáticos/farmacologia , Hipocampo/citologia , Hipocampo/metabolismo , Homeostase , Técnicas In Vitro , Indóis/farmacologia , Camundongos , Camundongos Knockout , Técnicas de Patch-Clamp , Fotólise , Tempo de Reação , Tapsigargina/farmacologia
17.
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
18.
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
20.
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
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA