Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Pregnancy Hypertens ; 22: 37-46, 2020 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-32721892

RESUMO

Oxidative stress and endothelial dysfunction contribute substantially to the pathogenesis of preeclampsia (PE). Decidual mesenchymal stem/stromal cells (DMSC), reportedly reduce endothelial cell dysfunction and alleviate PE-like symptoms in a murine model. However, as a therapeutic strategy, the use of whole DMSC presents significant technical limitations, which may be overcome by employing DMSC-secreted extracellular vesicles (DMSC_EV). DMSC_EV restoration of endothelial dysfunction through a paracrine effect may alleviate the clinical features of PE. OBJECTIVE: To determine whether DMSC-secreted, extracellular vesicles (DMSC_EV) restore endothelial cell function and reduce oxidative stress. METHODS: DMSC were isolated from the placentae of uncomplicated term pregnancies and DMSC_EV prepared by ultracentrifugation. Human umbilical vein endothelial cells (HUVEC) were treated with bacterial lipopolysaccharide (LPS), or with serum from PE patients, to model the effects of PE. DMSC_EV were then added to treated HUVEC and their growth profiles, inflammatory state, and oxidative stress levels measured. RESULTS: DMSC_EV displayed characteristic features of extracellular vesicles. In both LPS- and PE serum-treatment models, addition of DMSC_EV significantly increased HUVEC cell attachment and proliferation, and significantly reduced production of pro-inflammatory cytokine IL-6. The addition of DMSC_EV to LPS-treated HUVEC had no significant effect on total antioxidant capacity, superoxide dismutase levels or on lipid peroxidation levels. In contrast, the addition of DMSC_EV to PE serum-treated HUVEC resulted in a significant reduction in levels of lipid peroxidation. CONCLUSION: Addition of DMSC_EV had beneficial effects in both LPS- and PE serum- treated HUVEC but the two treatment models to induce endothelial cell dysfunction showed differences. The LPS treatment of HUVEC model may not accurately model the endothelial cell dysfunction characteristic of PE. Human cell culture models of PE show that DMSC_EV improve endothelial cell dysfunction in PE, but testing in in vivo models of PE is required.


Assuntos
Células Endoteliais da Veia Umbilical Humana/metabolismo , Células-Tronco Mesenquimais/metabolismo , Estresse Oxidativo , Placenta/metabolismo , Pré-Eclâmpsia/metabolismo , Adulto , Animais , Estudos de Casos e Controles , Proliferação de Células , Células Cultivadas/metabolismo , Decídua , Feminino , Humanos , Camundongos , Gravidez
2.
Mol Hum Reprod ; 26(8): 636-651, 2020 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-32609359

RESUMO

Ageing and parturition share common pathways, but their relationship remains poorly understood. Decidual cells undergo ageing as parturition approaches term, and these age-related changes may trigger labour. Mesenchymal stem/stromal cells (MSCs) are the predominant stem cell type in the decidua. Stem cell exhaustion is a hallmark of ageing, and thus ageing of decidual MSCs (DMSCs) may contribute to the functional changes in decidual tissue required for term spontaneous labour. Here, we determine whether DMSCs from patients undergoing spontaneous onset of labour (SOL-DMSCs) show evidence of ageing-related functional changes compared with those from patients not in labour (NIL-DMSCs), undergoing Caesarean section. Placentae were collected from term (37-40 weeks of gestation), SOL (n = 18) and NIL (n = 17) healthy patients. DMSCs were isolated from the decidua basalis that remained attached to the placenta after delivery. DMSCs displayed stem cell-like properties and were of maternal origin. Important cell properties and lipid profiles were assessed and compared between SOL- and NIL-DMSCs. SOL-DMSCs showed reduced proliferation and increased lipid peroxidation, migration, necrosis, mitochondrial apoptosis, IL-6 production and p38 MAPK levels compared with NIL-DMSCs (P < 0.05). SOL- and NIL-DMSCs also showed significant differences in lipid profiles in various phospholipids (phosphatidylethanolamine, phosphatidylglycerol, phosphatidylinositol, phosphatidylserine), sphingolipids (ceramide, sphingomyelin), triglycerides and acyl carnitine (P < 0.05). Overall, SOL-DMSCs had altered lipid profiles compared with NIL-DMSCs. In conclusion, SOL-DMSCs showed evidence of ageing-related reduced functionality, accumulation of cellular damage and changes in lipid profiles compared with NIL-DMSCs. These changes may be associated with term spontaneous labour.


Assuntos
Células-Tronco Mesenquimais/metabolismo , Células Estromais/metabolismo , Apoptose/fisiologia , Movimento Celular/fisiologia , Decídua/citologia , Decídua/metabolismo , Feminino , Humanos , Interleucina-6/metabolismo , Trabalho de Parto , Peroxidação de Lipídeos/fisiologia , Células-Tronco Mesenquimais/citologia , Necrose/metabolismo , Gravidez , Células Estromais/citologia
3.
Cell Adh Migr ; 10(3): 310-21, 2016 05 03.
Artigo em Inglês | MEDLINE | ID: mdl-27070187

RESUMO

A successful pregnancy depends on the intricate and timely interactions of maternal and fetal cells. Placental extravillous cytotrophoblast invasion involves a cellular transition from an epithelial to mesenchymal phenotype. Villous cytotrophoblasts undergo a partial epithelial to mesenchymal transition (EMT) when differentiating into extravillous cytotrophoblasts and gain the capacity to migrate and invade. This review summarizes our current knowledge regarding known regulators of EMT in the human placenta, including the inducers of EMT, upstream transcription factors that control EMT and the downstream effectors, cell adhesion molecules and their differential expression and functions in pregnancy pathologies, preeclampsia (PE) and fetal growth restriction (FGR). The review also describes the research strategies that were used for the identification of the functional role of EMT targets in vitro. A better understanding of molecular pathways driven by placental EMT and further elucidation of signaling pathways underlying the developmental programs may offer novel strategies of targeted therapy for improving feto-placental growth in placental pathologies including PE and FGR.


Assuntos
Transição Epitelial-Mesenquimal/fisiologia , Placenta/metabolismo , Trofoblastos/citologia , Diferenciação Celular , Transição Epitelial-Mesenquimal/genética , Feminino , Retardo do Crescimento Fetal , Humanos , Placenta/patologia , Placentação/fisiologia , Pré-Eclâmpsia/genética , Pré-Eclâmpsia/patologia , Gravidez
4.
Cells Tissues Organs ; 193(1-2): 23-40, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21051859

RESUMO

Epithelial-mesenchymal transition (EMT) is a feature of migratory cellular processes in all stages of life, including embryonic development and wound healing. Importantly, EMT features cluster with disease states such as chronic fibrosis and cancer. The dissolution of the E-cadherin-mediated adherens junction (AJ) is a key preliminary step in EMT and may occur early or late in the growing epithelial tumour. This is a first step for tumour cells towards stromal invasion, intravasation, extravasation and distant metastasis. The AJ may be inactivated in EMT by directed E-cadherin cleavage; however, it is increasingly evident that the majority of AJ changes are transcriptional and mediated by an expanding group of transcription factors acting directly or indirectly to repress E-cadherin expression. A review of the current literature has revealed that these factors may regulate each other in a hierarchical pattern where Snail1 (formerly Snail) and Snail2 (formerly Slug) are initially induced, leading to the activation of Zeb family members, TCF3, TCF4, Twist, Goosecoid and FOXC2. Within this general pathway, many inter-regulatory relationships have been defined which may be important in maintaining the EMT phenotype. This may be important given the short half-life of Snail1 protein. We have investigated these inter-regulatory relationships in the mesenchymal breast carcinoma cell line PMC42 (also known as PMC42ET) and its epithelial derivative, PMC42LA. This review also discusses several newly described regulators of E-cadherin repressors including oestrogen receptor-α and new discoveries in hypoxia- and growth factor-induced EMT. Finally, we evaluated how these findings may influence approaches to current cancer treatment.


Assuntos
Caderinas/metabolismo , Transição Epitelial-Mesenquimal/fisiologia , Junções Aderentes/genética , Junções Aderentes/metabolismo , Animais , Caderinas/genética , Linhagem Celular Tumoral , Transição Epitelial-Mesenquimal/genética , Receptor alfa de Estrogênio/genética , Receptor alfa de Estrogênio/metabolismo , Humanos , Hipóxia/genética , Hipóxia/fisiopatologia
5.
Dev Biol ; 321(2): 420-33, 2008 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-18652817

RESUMO

Recent studies implicate Wnt/beta-catenin signaling in lens differentiation (Stump, R. J., et al., 2003. A role for Wnt/beta-catenin signaling in lens epithelial differentiation. Dev Biol;259:48-61). Beta-catenin is a component of adherens junctions and functions as a transcriptional activator in canonical Wnt signaling. We investigated the effects of Cre/LoxP-mediated deletion of beta-catenin during lens development using two Cre lines that specifically deleted beta-catenin in whole lens or only in differentiated fibers, from E13.5. We found that beta-catenin was required in lens epithelium and during early fiber differentiation but appeared to be redundant in differentiated fiber cells. Complete loss of beta-catenin resulted in an abnormal and deficient epithelial layer with loss of E-cadherin and Pax6 expression as well as abnormal expression of c-Maf and p57(kip2) but not Prox1. There was also disrupted fiber cell differentiation, characterized by poor cell elongation, decreased beta-crystallin expression, epithelial cell cycle arrest at G(1)-S transition and premature cell cycle exit. Despite cell cycle arrest there was no induction of apoptosis. Mutant fiber cells displayed altered apical-basal polarity as evidenced by altered distribution of the tight junction protein, ZO1, disruption of apical actin filaments and abnormal deposition of extracellular matrix, resulting in a deficient lens capsule. Loss of beta-catenin also affected the formation of adhesion junctions as evidenced by dissociation of N-cadherin and F-actin localization in differentiating fiber cells. However, loss of beta-catenin from terminally differentiating fibers had no apparent effects on adhesion junctions between adjacent embryonic fibers. These data indicate that beta-catenin plays distinct functions during lens fiber differentiation and is involved in both Wnt signaling and adhesion-related mechanisms that regulate lens epithelium and early fiber differentiation.


Assuntos
Junções Aderentes/metabolismo , Células Epiteliais/metabolismo , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Cristalino/embriologia , Transdução de Sinais/fisiologia , Proteínas Wnt/metabolismo , beta Catenina/metabolismo , Animais , Diferenciação Celular/fisiologia , Polaridade Celular/fisiologia , Camundongos
6.
Cells Tissues Organs ; 185(1-3): 191-203, 2007.
Artigo em Inglês | MEDLINE | ID: mdl-17587825

RESUMO

Breast cancer is a highly prevalent disease among women worldwide. While the expression of certain proteins within these tumours is used for prognosis and selection of therapies, there is a continuing need for additional markers to be identified. A considerable amount of current literature, based predominantly on cell culture systems, suggests that a major mechanism responsible for the progression of breast cancer is due to tumour cells losing their epithelial features and gaining mesenchymal properties. These events are proposed to be very similar to the epithelial-mesenchymal transition (EMT) process that has been well characterised in embryonic development. For the developmental and putative cancer EMT, the cell intermediate filament status changes from a keratin-rich network which connects to adherens junctions and hemidesmosomes, to a vimentin-rich network connecting to focal adhesions. This review summarises observations of vimentin expression in breast cancer model systems, and discusses the potential role of EMT in human breast cancer progression, and the prognostic usefulness of vimentin expression.


Assuntos
Neoplasias da Mama/patologia , Epitélio/patologia , Mesoderma/patologia , Vimentina/metabolismo , Animais , Neoplasias da Mama/metabolismo , Linhagem Celular Tumoral , Epitélio/metabolismo , Feminino , Humanos , Imuno-Histoquímica , Técnicas In Vitro , Mesoderma/metabolismo , Camundongos , Camundongos SCID , Modelos Biológicos , Transplante de Neoplasias , Transplante Heterólogo , Vimentina/genética
7.
Mol Vis ; 13: 418-30, 2007 Mar 26.
Artigo em Inglês | MEDLINE | ID: mdl-17417603

RESUMO

PURPOSE: Regulation of lens development involves an intricate interplay between growth factor (e.g. FGF and TGFbeta) and extracellular matrix (ECM) signaling pathways. Focal adhesion kinase (FAK) is a cytoplasmic tyrosine kinase that plays key roles in transmitting ECM signals by integrins. In this study, we delineated patterns of FAK expression and tyrosine phosphorylation (Y397) in the developing lens and investigated its regulation by FGF2. We also examined FAK expression and activation during disrupted fiber differentiation in mice expressing a dominant-negative TGFbeta receptor. METHODS: FAK expression and activation (phosphorylation on Y397) was studied in embryonic and postnatal rodent lenses by in situ hybridization, immunofluorescence, and western blotting. Rat lens explants were used to investigate the effects of FGF2 on FAK expression and activation. Immunofluorescence and western blotting were used to examine FAK expression and phosphorylation in transgenic mice that express a dominant-negative TGFbeta receptor. RESULTS: FAK is widely expressed and phosphorylated during embryonic stages of lens morphogenesis and differentiation. However, in postnatal lenses its expression and activation becomes restricted to the posterior germinative zone and the transitional zone at the lens equator. While both NH2- and COOH-terminal antibodies revealed cytoplasmic and membrane-associated staining in lens cells, the NH2-terminal antibody also showed FAK was present in fiber cell nuclei. In vitro, FAK expression and phosphorylation on Y397 were increased by concentrations of FGF2 that initiate lens epithelial cell migration (10 ng/ml) and differentiation (50 ng/ml) but not proliferation (5 ng/ml). Moreover, reactivity for Y397 phosphorylated FAK is prominent in the nuclei of differentiating fibers both in vivo and in vitro. Disruption of TGFbeta-like signals by ectopic expression of a dominant-negative TGFbeta receptor (TbetaRII(D/N)) results in abnormal lens fiber differentiation in transgenic mice. While FAK expression is initiated normally in the posterior germinative zone of TbetaRII(D/N) transgenic lenses, as fiber differentiation proceeds, FAK becomes localized to a perinuclear compartment, decreases its association with the cytoskeleton and is poorly phosphorylated on Y(397). CONCLUSIONS: FAK is widely expressed and activated during early lens morphogenesis. During secondary lens fiber differentiation, FAK is expressed and phosphorylated on Y397 as epithelial cells exit the cell cycle, initiate migration at the equator, and undergo differentiation in the transitional zone. During terminal fiber differentiation an NH2-terminal fragment of FAK, including Y397, is translocated to the nucleus. The expression, activation, and nuclear localization of FAK are regulated, at least partly, by FGF2. FAK activity and subcellular localization are also modulated by TGFbeta-like signals. In fiber cells of TbetaRII(D/N) transgenic lenses, FAK is abnormally retained in a perinuclear compartment, loses its association with the cytoskeleton, and is poorly phosphorylated. These data suggest that integrin signaling via FAK plays important roles during lens differentiation, mediated by FGFs and TGFbeta-superfamily signals.


Assuntos
Envelhecimento/metabolismo , Proteína-Tirosina Quinases de Adesão Focal/metabolismo , Cristalino/embriologia , Cristalino/crescimento & desenvolvimento , Animais , Animais Recém-Nascidos , Embrião de Mamíferos/enzimologia , Embrião de Mamíferos/metabolismo , Ativação Enzimática , Fator 2 de Crescimento de Fibroblastos/metabolismo , Proteína-Tirosina Quinases de Adesão Focal/genética , Cristalino/enzimologia , Cristalino/metabolismo , Camundongos , Camundongos Transgênicos , Fosforilação , Proteínas Serina-Treonina Quinases , RNA Mensageiro/metabolismo , Ratos , Ratos Wistar , Receptor do Fator de Crescimento Transformador beta Tipo II , Receptores de Fatores de Crescimento Transformadores beta/genética , Distribuição Tecidual
8.
Gynecol Oncol ; 105(2): 462-9, 2007 May.
Artigo em Inglês | MEDLINE | ID: mdl-17300833

RESUMO

OBJECTIVES: Inhibition of cyclin-dependent-kinases (CDKs) represents an interesting approach in cancer therapy. We have explored this in cell lines of human uterine sarcoma-tumours associated with poor survival, chemo-unresponsiveness and deregulation of cell cycle components. We studied the effects of the CDK inhibitor seliciclib (CYC202, R-roscovitine) when used alone or in combination with cisplatin. METHODS: Cell lines used: SK-UT-1, SK-UT-1b and SK-LMS-1, the cytotoxicity of seliciclib and cisplatin was measured by the MTT assay. In combination with cisplatin the effects of seliciclib were examined by isobologram analysis. CDK2 levels were examined at mRNA and protein level by immunoblotting and PCR. We also looked at the effects of seliciclib on p53-dependent response of cells to seliciclib using immunoblotting. The effects of combination treatment were analysed using annexin V and PI staining by flow cytometric analysis. RESULTS: IC50 values for seliciclib were 10.5, 7.1 and 25.7 microM, for SK-UT-1, SK-UT-1b and SK-LMS-1 respectively, P53 in the SK-UT-1b (wild-type) and SK-LMS-1 lines (mutant) showed a wild-type response with induction seen with seliciclib treatment for 24 and 48 h. Seliciclib caused a decrease in CDK2 mRNA and protein over 72 h. A combination of cisplatin and seliciclib was synergistic in all three cell lines. Effects of combination treatment were an enhancement in apoptosis as judged by the emergence of a sub-G1 population in cell cycle analysis and a sub-G1 population with PI staining. CONCLUSIONS: Our data demonstrate the effectiveness of seliciclib as a single agent and when used in combination with cisplatin where the effects are synergistic.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Quinases Ciclina-Dependentes/antagonistas & inibidores , Inibidores de Proteínas Quinases/farmacologia , Purinas/farmacologia , Sarcoma/tratamento farmacológico , Neoplasias Uterinas/tratamento farmacológico , Anexina A5 , Apoptose/efeitos dos fármacos , Cisplatino/administração & dosagem , Quinase 2 Dependente de Ciclina/biossíntese , Quinase 2 Dependente de Ciclina/genética , Inibidor de Quinase Dependente de Ciclina p21/biossíntese , Inibidor de Quinase Dependente de Ciclina p21/genética , Inibidor de Quinase Dependente de Ciclina p27 , Feminino , Citometria de Fluxo , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/genética , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Inibidores de Proteínas Quinases/administração & dosagem , Purinas/administração & dosagem , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , Roscovitina , Sarcoma/genética , Sarcoma/metabolismo , Sarcoma/patologia , Ativação Transcricional , Neoplasias Uterinas/genética , Neoplasias Uterinas/metabolismo , Neoplasias Uterinas/patologia
9.
Mol Vis ; 10: 566-76, 2004 Aug 23.
Artigo em Inglês | MEDLINE | ID: mdl-15346106

RESUMO

PURPOSE: Members of the TGFbeta super-family have been shown to play important roles in lens development, including lens placode formation and fiber differentiation, and also induce changes characteristic of some forms of cataract. Previous studies demonstrated expression of TGFbeta receptors during lens morphogenesis. However, the expression patterns of activin and BMP receptors or their signaling mediators, the Smad proteins, have not been well documented. In this study we examine the spatio-temporal expression patterns of activin receptors (ActRIIA, ActRIIB, ALK1, and ALK2), BMP receptors (BMPRII, ALK3, and ALK6), and the distribution of the phosphorylated forms of Smad1 and Smad2 during normal lens development (E12-P21) and aberrant development in transgenic mice that express dominant negative TGFbeta receptors. METHODS: RT-PCR was used to identify receptor expression in total RNA isolated from P2 and P21 rat lenses. cDNAs were cloned and used for in situ hybridization analysis of spatio-temporal expression patterns in wild type and transgenic (OVE550 and OVE591) lenses. Expression of ALK3 was also examined by immunofluorescence and immunoblotting. Antibodies for phosphorylated forms of Smad1 and Smad2 were used to examine activation of BMP and activin signaling. RESULTS: RT-PCR of RNA from postnatal lenses showed distinct expression of ActRIIA, ActRIIB, BMPRII, and ALK3 but not ALK1, ALK2, or ALK 6. In situ hybridization with specific probes for BMPRII, ActRII, and ALK3 showed ubiquitous expression in ectoderm, lens pit, optic vesicle, and peri-optic mesenchyme during early lens formation at E12. During subsequent lens differentiation, from E14 onwards, expression of these receptors became increasingly restricted to the lens epithelium and to the equatorial region, including the germinative and transitional zones, where cells proliferate and commence differentiation, respectively. Expression for both receptors declined rapidly with fiber differentiation and maturation. Immunofluorescence with specific antibodies for phospho-Smad1 and phospho-Smad2 showed distinct localization of these signaling mediators in epithelial cells of the germinative zone and in fibers undergoing early differentiation in the transitional zone. Further investigation of the expression of these receptors in lenses of transgenic mice, which ectopically express a truncated TbetaRII, showed marked up regulation and aberrant expression of ALK3, but not BMPRII or ActRII. CONCLUSIONS: These results indicate that multiple members of the TGFbeta family have the potential to signal during lens fiber differentiation and suggest there may be cross-talk between different signaling pathways.


Assuntos
Receptores de Ativinas/genética , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Cristalino/embriologia , Receptores de Fatores de Crescimento/genética , Receptores de Ativinas/metabolismo , Animais , Western Blotting , Receptores de Proteínas Morfogenéticas Ósseas , Diferenciação Celular , Sondas de DNA/química , DNA Complementar/genética , Proteínas de Ligação a DNA/metabolismo , Técnica Indireta de Fluorescência para Anticorpo , Hibridização In Situ , Cristalino/citologia , Cristalino/crescimento & desenvolvimento , Cristalino/metabolismo , Camundongos , Camundongos Transgênicos , Morfogênese , RNA Mensageiro/metabolismo , Ratos , Ratos Wistar , Receptores de Fatores de Crescimento/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais , Proteínas Smad , Proteína Smad1 , Proteína Smad2 , Transativadores/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...