Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 63
Filtrar
1.
Cells ; 13(2)2024 01 06.
Artículo en Inglés | MEDLINE | ID: mdl-38247805

RESUMEN

Low-dose aspirin (LDA) is efficacious in preventing preeclampsia, but its mechanism of action is unclear. Conflicting evidence suggests that it may inhibit placental trophoblast release of soluble fms-like tyrosine kinase-1 (sFlt1), a key mediator of preeclampsia. We examined whether, and at what concentrations, aspirin and its principal metabolite, salicylic acid, modulate sFlt1 release and/or expression in trophoblasts. Human trophoblast lines BeWo and HTR-8/SVneo were cultured; BeWo cells were also treated with 1% oxygen vs. normoxia to mimic hypoxia in preeclamptic placentas. Cells were treated with aspirin or salicylic acid vs. vehicle for 24 h at concentrations relevant to LDA and at higher concentrations. Protein concentrations (ELISA) and mRNA expression (RT-PCR) of sFlt1 were determined. Under normoxia, LDA-relevant concentrations of aspirin (10-50 µmol/L) or salicylic acid (20-100 µmol/L) had no significant effect on sFlt1 protein release or mRNA expression in BeWo cells. However, inhibition was observed at higher concentrations (1 mmol/L for aspirin and ≥200 µmol/L for salicylic acid). Hypoxia enhanced sFlt1 protein release and mRNA expression in BeWo cells, but these responses were not significantly affected by either aspirin or salicylic acid at LDA concentrations. Similarly, neither drug altered sFlt1 protein secretion or mRNA expression in normoxic HTR-8/SVneo cells at LDA concentrations. We suggest that direct modulation of trophoblast release or expression of sFlt1 is unlikely to be a mechanism underlying the clinical efficacy of LDA in preeclampsia.


Asunto(s)
Aspirina , Preeclampsia , Trofoblastos , Receptor 1 de Factores de Crecimiento Endotelial Vascular , Femenino , Humanos , Embarazo , Aspirina/farmacología , Hipoxia , Placenta , Preeclampsia/tratamiento farmacológico , Proteínas Tirosina Quinasas Receptoras , ARN Mensajero/genética , Ácido Salicílico/farmacología , Trofoblastos/efectos de los fármacos , Trofoblastos/metabolismo , Receptor 1 de Factores de Crecimiento Endotelial Vascular/efectos de los fármacos
2.
Pregnancy Hypertens ; 26: 127-132, 2021 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-34773881

RESUMEN

Disruption of well-controlled reproductive functions leads to pregnancy complications such as hypertensive disorders of pregnancy (HDP). Uncaria tomentosa (Wild), known as cat's claw, is widely used for the treatment of a various types of health problems; AC-11 (AC-11®, hot-water extract of U. tomentosa) is unique phytochemical compound and has potential roles as anti-inflammatory or anti-oxidant processes. We investigated whether AC-11 has a protective effect on pathogenesis of HDP in vivo and production of anti-angiogenic factors (sFlt-1 and sEng, major factors for the onset of HDP) in in vitro. Non-pregnant or pregnant mice were administered AC-11 (4 mg/mL), then, angiotensin II (Ang II) was subcutaneously infused to increase blood pressure. Human placental tissues or human umbilical vein endothelial cells (HUVECs) were incubated with or without AC-11. Treatment with AC-11 significantly reduced blood pressure induced by Ang II infusion. The population of CD8+T cells, the ratio of CD8/CD4, and plasma interleukin-6 levels were increased by Ang II infusion, and were decreased by AC-11 both in pregnant and non-pregnant mice. In pregnant mice, plasma levels of sFlt-1 and sEng were decreased by AC-11. In in vitro cell culture of HUVECs or placental tissue culture, treatment with AC-11 significantly inhibited secretion of sFlt-1 and sEng. We suggest a novel role of AC-11 in regulating blood pressure by controlling the balance of T cell population and inflammatory cytokine production both in non-pregnant and pregnant conditions. In addition, AC-11 inhibits HDP-related factors, including sFlt-1 and sEng, suggesting that AC-11 may useful for relieving HDP.


Asunto(s)
Presión Sanguínea/efectos de los fármacos , Uña de Gato , Extractos Vegetales/farmacología , Preeclampsia/metabolismo , Animales , Endoglina/efectos de los fármacos , Femenino , Humanos , Ratones , Extractos Vegetales/administración & dosificación , Preeclampsia/prevención & control , Embarazo , Receptor 1 de Factores de Crecimiento Endotelial Vascular/efectos de los fármacos
3.
Pregnancy Hypertens ; 22: 86-92, 2020 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-32758704

RESUMEN

Development and repurposing of therapies that show promise in the prevention or treatment of preeclampsia would be a major advance for the obstetrics field. We recently identified esomeprazole and sulfasalazine as potential candidates for the treatment of preeclampsia. Both reduce placental and endothelial secretion of sFlt-1 and sENG and mitigate endothelial dysfunction in vitro. Here we assessed whether esomeprazole and sulfasalazine in combination would additively attenuate the elevated release of anti-angiogenic factors and markers of endothelial dysfunction, key characteristics of preeclampsia. Primary placental tissue and cells, and primary endothelial cells were treated with esomeprazole and sulfasalazine alone and in combination. We assessed secretion of sFlt-1 and sENG and performed in vitro assays of endothelial dysfunction. Combining esomeprazole and sulfasalazine in lower concentrations caused an additive reduction in sFlt-1 secretion in primary cytotrophoblasts, placental explants and endothelial cells. No additive reduction was observed in sENG secretion when esomeprazole and sulfasalazine were combined. Together, esomeprazole and sulfasalazine additively reduced TNF-α-induced VCAM and ET-1 mRNA expression, and monocyte adhesion to endothelial cells. In conclusion, combining esomeprazole and sulfasalazine additively reduced secretion of sFlt-1 and markers of endothelial dysfunction. Combined administration of esomeprazole and sulfasalazine may provide a more effective treatment or prevention for preeclampsia compared to either as single agents.


Asunto(s)
Esomeprazol/farmacología , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Preeclampsia/metabolismo , Sulfasalazina/farmacología , Receptor 1 de Factores de Crecimiento Endotelial Vascular/efectos de los fármacos , Estudios de Casos y Controles , Quimioterapia Combinada , Femenino , Humanos , Placenta/efectos de los fármacos , Embarazo
4.
BJOG ; 127(4): 478-488, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-31715077

RESUMEN

OBJECTIVE: Women with pre-eclampsia have elevated circulating levels of soluble fms-like tyrosine kinase-1 (sFlt-1). Statins can reduce sFlt-1 from cultured cells and improve pregnancy outcome in animals with a pre-eclampsia-like syndrome. We investigated the effect of pravastatin on plasma sFlt-1 levels during pre-eclampsia. DESIGN: Blinded (clinician and participant), proof of principle, placebo-controlled trial. SETTING: Fifteen UK maternity units. POPULATION: We used a minimisation algorithm to assign 62 women with early-onset pre-eclampsia (24+0 -31+6  weeks of gestation) to receive pravastatin 40 mg daily (n = 30) or matched placebo (n = 32), from randomisation to childbirth. PRIMARY OUTCOME: Difference in mean plasma sFlt-1 levels over the first 3 days following randomisation. RESULTS: The difference in the mean maternal plasma sFlt-1 levels over the first 3 days after randomisation between the pravastatin (n = 27) and placebo (n = 29) groups was 292 pg/ml (95% CI -1175 to 592; P = 0.5), and over days 1-14 was 48 pg/ml (95% CI -1009 to 913; P = 0.9). Women who received pravastatin had a similar length of pregnancy following randomisation compared with those who received placebo (hazard ratio 0.84; 95% CI 0.50-1.40; P = 0.6). The median time from randomisation to childbirth was 9 days (interquartile range [IQR] 5-14 days) for the pravastatin group and 7 days (IQR 4-11 days) for the placebo group. There were three perinatal deaths in the placebo-treated group and no deaths or serious adverse events attributable to pravastatin. CONCLUSIONS: We found no evidence that pravastatin lowered maternal plasma sFlt-1 levels once early-onset pre-eclampsia had developed. Pravastatin appears to have no adverse perinatal effects. TWEETABLE ABSTRACT: Pravastatin does not improve maternal plasma sFlt-1 or placental growth factor levels following a diagnosis of early preterm pre-eclampsia #clinicaltrial finds.


Asunto(s)
Inhibidores de Hidroximetilglutaril-CoA Reductasas/administración & dosificación , Pravastatina/administración & dosificación , Preeclampsia/tratamiento farmacológico , Receptor 1 de Factores de Crecimiento Endotelial Vascular/sangre , Adulto , Método Doble Ciego , Femenino , Edad Gestacional , Humanos , Preeclampsia/sangre , Embarazo , Receptor 1 de Factores de Crecimiento Endotelial Vascular/efectos de los fármacos
5.
Pregnancy Hypertens ; 14: 125-130, 2018 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-30527099

RESUMEN

Preeclampsia is associated with intermittent placental hypoxia, inflammation and the release of antiangiogenic factors, namely sFLT-1 and sEng. These factors cause maternal endothelial dysfunction and the manifestation of clinical disease. Disulfiram is a dehydrogenase inhibitor used to treat alcoholism and has been suggested as a proteasome inhibitor. Inhibiting the proteasome has been previously shown to reduce FLT-1 gene expression. Thus, we aim to investigate whether disulfiram alters the secretion of sFLT-1 and sEng and reduces endothelial dysfunction. METHODS AND RESULTS: We assessed the effects of disulfiram on primary cytotrophoblast and human umbilical vein endothelial cells (HUVECs). Disulfiram significantly reduced mRNA expression of membrane bound FLT-1 and sFLT-1 variants in primary cytotrophoblasts, which translated into a significant reduction in the protein secretion of sFLT-1. Additionally, sFLT-1 was reduced in primary HUVECs treated with disulfiram, whilst sEng was only reduced in primary cytotrophoblasts. Next, we investigated the effect of disulfiram on endothelial dysfunction using primary HUVECs treated with 5% preeclamptic serum ±â€¯disulfiram. Serum from preeclamptic women induced endothelial dysfunction evidenced by increased mRNA expression of vascular cell adhesion molecule-1 (VCAM-1) and adhesion of peripheral blood mononuclear cells (PBMCs) to HUVECs. The addition of disulfiram reduced VCAM-1 mRNA expression, however did not affect the adhesion of PBMCs to endothelial cells. Lastly, we assessed the effects of disulfiram on the 20S subunit of the proteasome and found disulfiram did not inhibit this subunit in either primary cytotrophoblast or HUVECs. CONCLUSIONS: Disulfiram quenches sFLT-1 and sEng via mechanisms independent of the 20S subunit of the proteasome. Understanding of the mechanisms by which disulfiram inhibits antiangiogenic secretion may reveal insights into the pathogenesis and potential therapeutic targets for preeclampsia.


Asunto(s)
Inhibidores del Acetaldehído Deshidrogenasa/farmacología , Disulfiram/farmacología , Endoglina/efectos de los fármacos , Placenta/efectos de los fármacos , Preeclampsia/metabolismo , Receptor 1 de Factores de Crecimiento Endotelial Vascular/efectos de los fármacos , Endoglina/genética , Endoglina/metabolismo , Ensayo de Inmunoadsorción Enzimática , Femenino , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Humanos , Placenta/metabolismo , Embarazo , Complejo de la Endopetidasa Proteasomal/efectos de los fármacos , ARN Mensajero/metabolismo , Receptor 1 de Factores de Crecimiento Endotelial Vascular/genética , Receptor 1 de Factores de Crecimiento Endotelial Vascular/metabolismo
6.
Pregnancy Hypertens ; 13: 1-6, 2018 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-30177033

RESUMEN

OBJECTIVES: To examine the effect of sildenafil on level of antiangiogenic proteins of preeclampsia. Firstly to examine the effect of sildenafil on serum biomarkers in a patient with preterm preeclampsia. Secondly, to examine the effect of sildenafil on sFlt-1 and soluble endoglin secretion from primary trophoblasts and placental explants. STUDY DESIGN: The clinical team administered 50 mg tds sildenafil to a 26-year-old primigravid woman with severe preeclampsia at the threshold of viability (24 3/7 weeks gestation) and we collected bloods to examine the effect of slidenafil on antiangiogenic factors sFlt-1 and soluble endoglin (sENG), pro-angiogenic factor PlGF and vascular cell adhesion molecule 1 (VCAM-1) and endothelin-1 (ET1). We administered sildenafil to human primary trophoblasts and placental explants and explored its effect on sFlt-1 and sENG secretion. MAIN OUTCOME MEASURES: We examined serum anti-angiogenic factors sFlt-1 and sENG, pro-angiogenic factor PlGF, the potent vasoconstrictor ET1 and VCAM-1 by ELISA. We explored the effect of sildenafil on sFlt-1 secretion from primary trophoblasts and sFlt-1 and sENG secretion from placental explants. RESULTS: We found a reduction in serum sFlt-1, stabilisation in sENG and PlGF in a patient with peri-viable preterm preeclampsia administered oral sildenafil 50 mg three times daily (tds). Furthermore there was an initial stabilisation in serum VCAM-1 and a decline in ET1 with sildenafil administration. This was concordant with stabilisation of clinical and biochemical features of preeclampsia. Interestingly, treating placental cells and tissues in vitro with sildenafil did not appear to change sFlt-1 or sENG secretion. CONCLUSION: Sildenafil administration was associated with a reduction in serum sFlt-1 and sENG secretion and increase in PlGF secretion in a patient with preterm preeclampsia, potentially via increasing placental perfusion rather than acting directly on the placenta.


Asunto(s)
Preeclampsia/sangre , Citrato de Sildenafil/farmacología , Receptor 1 de Factores de Crecimiento Endotelial Vascular/efectos de los fármacos , Vasodilatadores/farmacología , Administración Oral , Adulto , Biomarcadores/sangre , Femenino , Humanos , Placenta/efectos de los fármacos , Embarazo , Citrato de Sildenafil/administración & dosificación , Trofoblastos/efectos de los fármacos , Receptor 1 de Factores de Crecimiento Endotelial Vascular/sangre , Vasodilatadores/administración & dosificación
7.
World Neurosurg ; 120: e380-e391, 2018 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-30144594

RESUMEN

BACKGROUND: Glioblastoma (GBM) is the most common and fatal human brain tumor, with the worst prognosis. The aberrant microenvironment, enhanced by the activation of proangiogenic mediators such as hypoxia-inducible factor-1α (HIF-1α), vascular endothelial growth factor (VEGF), and their downstream effectors, sustain GBM malignancy. Proangiogenic signaling represents an attractive chemotherapeutic target. Recent evidence suggests a therapeutic benefit from aspirin (acetylsalicylic acid, or ASA) intake in reducing risk and cancer progression. METHODS: In the present study, human primary GBM-endothelial cells (ECs) were used to ascertain whether ASA could inhibit angiogenesis and improve cell sensitivity to drugs. The impact of ASA was observed by measuring cell viability, tube-like structure formation, migration, VEGF production, and proliferative, proangiogenic, and apoptotic modulators expression, such as HIF-1α/VEGF/vascular endothelial growth factor receptor/(VEGFR)-1/VEGFR-2, Ras/mitogen-activated protein kinase kinase/extracellular signal-regulated kinase, phosphoinositide 3-kinase/AKT signaling axis, and Bcl-2-associated X protein/B-cell lymphoma 2 (BCL-2) ratio. Furthermore, we evaluated the effect of ASA alone or in combination with temozolomide (TMZ), bevacizumab (BEV), and sunitinib (SUN). RESULTS: Our data reported that ASA affected GBM-EC viability, tube-like structure formation, cell migration, and VEGF releasing in a dose-dependent manner and that combined treatments with TMZ, BEV, and SUN synergized to counteract proangiogenic cell ability. mRNA expression analysis displayed a marked effect of ASA in reducing VEGF, VEGFR-1, HIF-1α, RAS, mitogen-activated protein kinase kinase, AKT, and BCL-2, as well a combined anticancer effect of ASA together with TMZ, BEV, and SUN. Levels of HIF-1α, VEGFR-2, Bcl-2-associated X protein, and BCL-2 protein expression confirmed a positive trend. CONCLUSIONS: ASA and antiangiogenic therapies showed synergetic anticancer efficacy in human primary GBM-ECs. Thus, the combination of conventional chemotherapy with ASA may offer a new strategy to counteract tumor malignancy.


Asunto(s)
Antiinflamatorios no Esteroideos/farmacología , Antineoplásicos/farmacología , Aspirina/farmacología , Bevacizumab/farmacología , Neoplasias Encefálicas/irrigación sanguínea , Células Endoteliales/efectos de los fármacos , Glioblastoma/irrigación sanguínea , Sunitinib/farmacología , Temozolomida/farmacología , Inhibidores de la Angiogénesis/farmacología , Antineoplásicos Alquilantes/farmacología , Antineoplásicos Inmunológicos/farmacología , Western Blotting , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/metabolismo , Supervivencia Celular/efectos de los fármacos , Sinergismo Farmacológico , Células Endoteliales/metabolismo , Glioblastoma/genética , Glioblastoma/metabolismo , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/efectos de los fármacos , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Quinasas de Proteína Quinasa Activadas por Mitógenos/efectos de los fármacos , Quinasas de Proteína Quinasa Activadas por Mitógenos/genética , Quinasas de Proteína Quinasa Activadas por Mitógenos/metabolismo , Proteínas Proto-Oncogénicas c-akt/efectos de los fármacos , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proteínas Proto-Oncogénicas c-bcl-2/efectos de los fármacos , Proteínas Proto-Oncogénicas c-bcl-2/genética , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , ARN Mensajero/efectos de los fármacos , ARN Mensajero/metabolismo , Reacción en Cadena en Tiempo Real de la Polimerasa , Transducción de Señal , Factor A de Crecimiento Endotelial Vascular/efectos de los fármacos , Factor A de Crecimiento Endotelial Vascular/genética , Factor A de Crecimiento Endotelial Vascular/metabolismo , Receptor 1 de Factores de Crecimiento Endotelial Vascular/efectos de los fármacos , Receptor 1 de Factores de Crecimiento Endotelial Vascular/genética , Receptor 1 de Factores de Crecimiento Endotelial Vascular/metabolismo , Receptor 2 de Factores de Crecimiento Endotelial Vascular/efectos de los fármacos , Receptor 2 de Factores de Crecimiento Endotelial Vascular/genética , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo , Proteína X Asociada a bcl-2/efectos de los fármacos , Proteína X Asociada a bcl-2/genética , Proteína X Asociada a bcl-2/metabolismo , Proteínas ras/efectos de los fármacos , Proteínas ras/genética , Proteínas ras/metabolismo
8.
Biol Reprod ; 99(5): 1082-1090, 2018 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-29860275

RESUMEN

Low molecular weight heparin (LMWH) is being investigated as a potential preventative therapy against preeclampsia. There is evidence suggesting that LMWH may prevent preeclampsia through anticoagulation-independent mechanisms. In this study, we compared the in vitro placental, endothelial, and anti-inflammatory effects of an LMWH (dalteparin) with a nonanticoagulant, glycol-split heparin derivative (gsHep). In contrast with dalteparin, gsHep did not interact with antithrombin III, possess significant anti-Factor Xa activity, or significantly prolong in vitro plasma clotting time. However, dalteparin and gsHep were otherwise mechanistically similar, both interacting with soluble fms-like tyrosine kinase-1 (sFlt1) and promoting release of the pro-angiogenic protein placental growth factor, but not the antiangiogenic sFlt1, from healthy placental villous explants. Placental explant media pretreated with dalteparin or gsHep significantly stimulated endothelial cell tube formation compared to untreated explants. Lastly, dalteparin and gsHep both significantly suppressed inflammation by inhibiting complement activation and leukocyte adhesion to endothelial cells that were activated using serum from preeclamptic women. Our data suggest that nonanticoagulant heparin derivatives may be utilized as a tool to distinguish the anticoagulation-independent mechanisms of LMWH, and provide insight into the role of anticoagulation in the prevention of preeclampsia.


Asunto(s)
Antiinflamatorios no Esteroideos/farmacología , Glicoles/química , Heparina de Bajo-Peso-Molecular/farmacología , Células Endoteliales de la Vena Umbilical Humana/patología , Inflamación/fisiopatología , Placenta/patología , Preeclampsia/fisiopatología , Transducción de Señal/efectos de los fármacos , Anticoagulantes/farmacología , Adhesión Celular/efectos de los fármacos , Activación de Complemento/efectos de los fármacos , Dalteparina/farmacología , Factor Xa , Femenino , Heparina de Bajo-Peso-Molecular/química , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Humanos , Placenta/efectos de los fármacos , Embarazo , Receptor 1 de Factores de Crecimiento Endotelial Vascular/efectos de los fármacos , Receptor 1 de Factores de Crecimiento Endotelial Vascular/metabolismo
9.
Artículo en Inglés | MEDLINE | ID: mdl-29034214

RESUMEN

Malaria is a severe disease and kills over 400,000 people each year. Malarial complications are the main cause of death and include cerebral malaria and malaria-associated acute respiratory distress syndrome (MA-ARDS). Despite antimalarial treatment, lethality rates of MA-ARDS are still between 20 and 80%. Patients develop pulmonary edema with hemorrhages and leukocyte extravasation in the lungs. The vascular endothelial growth factor-A (VEGF-A) and the placental growth factor (PlGF) are vascular permeability factors and may be involved in the disruption of the alveolar-capillary membrane, leading to alveolar edema. We demonstrated increased pulmonary VEGF-A and PlGF levels in lungs of mice with experimental MA-ARDS. Depletion of pathogenic CD8+ T cells blocked pulmonary edema and abolished the increase of VEGF-A and PlGF. However, neutralization of VEGF receptor-2 (VEGFR-2) with the monoclonal antibody clone DC101 did not decrease pulmonary pathology. The broader spectrum receptor tyrosine kinase inhibitor sunitinib even increased lung pathology. These data suggest that the increase in alveolar VEGF-A and PlGF is not a cause but rather a consequence of the pulmonary pathology in experimental MA-ARDS and that therapeutic inhibition of VEGF receptors is not effective and even contra-indicated.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Malaria/complicaciones , Síndrome de Dificultad Respiratoria/etiología , Factor A de Crecimiento Endotelial Vascular/metabolismo , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo , Células Epiteliales Alveolares , Animales , Anticuerpos Monoclonales/inmunología , Anticuerpos Neutralizantes , Linfocitos T CD8-positivos/fisiología , Citocinas/metabolismo , Modelos Animales de Enfermedad , Edema/etiología , Femenino , Regulación de la Expresión Génica , Inmunoglobulina G/sangre , Inmunohistoquímica , Pulmón/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Factor de Crecimiento Placentario/metabolismo , Plasmodium berghei , Síndrome de Dificultad Respiratoria/tratamiento farmacológico , Síndrome de Dificultad Respiratoria/patología , Factor A de Crecimiento Endotelial Vascular/genética , Receptor 1 de Factores de Crecimiento Endotelial Vascular/efectos de los fármacos , Receptor 1 de Factores de Crecimiento Endotelial Vascular/inmunología , Receptor 1 de Factores de Crecimiento Endotelial Vascular/metabolismo , Receptor 2 de Factores de Crecimiento Endotelial Vascular/efectos de los fármacos , Receptor 2 de Factores de Crecimiento Endotelial Vascular/inmunología
10.
J Am Heart Assoc ; 5(7)2016 07 12.
Artículo en Inglés | MEDLINE | ID: mdl-27405812

RESUMEN

BACKGROUND: Inhibition of glycogen synthase kinase 3ß (GSK-3ß) has been reported to be cardioprotective during stressful conditions. METHODS AND RESULTS: Pigs were fed a high-fat diet for 4 weeks to develop metabolic syndrome, then underwent placement of an ameroid constrictor to their left circumflex artery to induce chronic myocardial ischemia. Two weeks later, animals received either: no drug (high cholesterol control group [HCC]) or a GSK-3ß inhibitor (GSK-3ß inhibited group [GSK-3ßI]), which were continued for 5 weeks, followed by myocardial tissue harvest. Coronary blood flow and vessel density were significantly increased in the GSK-3ßI group compared to the HCC group. Expression levels of the following proteins were greater in the GSK-3ßI group compared to the HCC group: vascular endothelial growth factor receptor 1 , vascular endothelial cadherin, γ-catenin, ß-catenin, protein kinase B, phosphorylated forkhead box O1, and superoxide dismutase 2. CONCLUSIONS: In the setting of metabolic syndrome, inhibition of GSK-3ß increases blood flow and vessel density in chronically ischemic myocardium. We identified several angiogenic, cell survival, and differentiation pathways that include ß-catenin signaling and AKT/FOXO1, through which GSK-3ß appears to improve vessel density and blood flow. These results may provide a potential mechanism for medical therapy of patients suffering from coronary artery disease and metabolic syndrome.


Asunto(s)
Vasos Coronarios/efectos de los fármacos , Glucógeno Sintasa Quinasa 3 beta/antagonistas & inhibidores , Corazón/efectos de los fármacos , Indoles/farmacología , Maleimidas/farmacología , Síndrome Metabólico/metabolismo , Isquemia Miocárdica/metabolismo , Miocardio/patología , Neovascularización Fisiológica/efectos de los fármacos , Animales , Cadherinas/efectos de los fármacos , Cadherinas/metabolismo , Enfermedad Crónica , Circulación Coronaria/efectos de los fármacos , Vasos Coronarios/patología , Dieta Alta en Grasa , Modelos Animales de Enfermedad , Proteína Forkhead Box O1/efectos de los fármacos , Proteína Forkhead Box O1/metabolismo , Isquemia Miocárdica/patología , Proteínas Proto-Oncogénicas c-akt/efectos de los fármacos , Proteínas Proto-Oncogénicas c-akt/metabolismo , Superóxido Dismutasa/efectos de los fármacos , Superóxido Dismutasa/metabolismo , Sus scrofa , Porcinos , Receptor 1 de Factores de Crecimiento Endotelial Vascular/efectos de los fármacos , Receptor 1 de Factores de Crecimiento Endotelial Vascular/metabolismo , beta Catenina/efectos de los fármacos , beta Catenina/metabolismo , gamma Catenina/efectos de los fármacos , gamma Catenina/metabolismo
11.
BMC Pregnancy Childbirth ; 16: 117, 2016 05 20.
Artículo en Inglés | MEDLINE | ID: mdl-27207105

RESUMEN

BACKGROUND: Preeclampsia is associated with the placental release of soluble fms-like tyrosine kinase 1 (sFlt-1) and soluble endoglin (sENG). These anti-angiogenic factors cause hypertension and multi-organ injury. Pravastatin decreases placental secretion of sFlt-1 in vitro and is currently being examined in clinical trials as a potential treatment for preeclampsia. However, it is possible that different classes of statins may be more potent at decreasing sFlt-1 secretion. We compared the relative potency of three different generations of statins on sFlt-1 and sENG secretion from human endothelial cells, trophoblast cells, and placenta explants. METHODS: We performed functional experiments using primary human umbilical vein endothelial cells, trophoblast cells and preterm preeclamptic placental explants to assess the affect of simvastatin, rosuvastatin and pravastatin on sFlt-1 and sENG secretion and compared the relative potency of each statin at reducing these factors (Inhibitory Concentration 50). Furthermore we assessed the effect of each statin on the antioxidant and cytoprotective enzyme, heme-oxygenase 1. RESULTS: All statins reduced sFlt-1 secretion from endothelial cells, trophoblasts and preterm preeclamptic placental explants. Simvastatin was the most potent inhibitor of sFlt-1 secretion from endothelial cells (IC 50 3.2 µM), trophoblast cells (IC 50 61.4 µM) and placental explants. Simvastatin was 28 times and 3 times more potent at reducing sFlt-1 secretion from endothelial cells and 85 times and 33 times more potent at reducing sFlt-1 secretion from trophoblast cells than pravastatin or rosuvastatin respectively. All statins increased sENG secretion from endothelial cells however did not change secretion from placental explants. While all statins up-regulated heme-oxygenase 1 in endothelial cells, only simvastatin up-regulated its expression in placenta from patients with preterm preeclampsia. CONCLUSION: Simvastatin may be a more potent inhibitor of sFlt-1 secretion from endothelial cells, trophoblast cells and placenta from women with preterm preeclampsia than either pravastatin or rosuvastatin.


Asunto(s)
Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Inhibidores de Hidroximetilglutaril-CoA Reductasas/farmacología , Placenta/efectos de los fármacos , Proteínas Gestacionales/efectos de los fármacos , Trofoblastos/efectos de los fármacos , Endoglina/efectos de los fármacos , Femenino , Hemo-Oxigenasa 1/efectos de los fármacos , Humanos , Placenta/metabolismo , Pravastatina/farmacología , Preeclampsia/tratamiento farmacológico , Embarazo , Proteínas Gestacionales/metabolismo , Rosuvastatina Cálcica/farmacología , Simvastatina/farmacología , Trofoblastos/citología , Regulación hacia Arriba , Receptor 1 de Factores de Crecimiento Endotelial Vascular/efectos de los fármacos , Receptor 1 de Factores de Crecimiento Endotelial Vascular/metabolismo
12.
Invest Ophthalmol Vis Sci ; 56(9): 5450-6, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-26284550

RESUMEN

PURPOSE: To determine the possible antiangiogenic effect of metalloproteinase (MMP) 14 cleavage of vascular endothelial growth factor receptor 1 (VEGFR1) in the cornea. METHODS: Recombinant mouse (rm) VEGFR1 was incubated with various concentrations of recombinant MMP14 to examine proteolysis in vitro. The reaction mixture was analyzed by SDS-PAGE and stained with Coomassie blue. The fragments resulting from rmVEGFR1 cleavage by MMP14 were subjected to Edman degradation, and the amino acid sequences were aligned with rmVEGFR1 sequences. Surface plasmon resonance was used to determine the equilibrium dissociation constant (KD) between MMP14 and rmVEGFR1. The KD value of rmVEGFR1 and the 59.8-kDa cleavage product binding to VEGF-A165 was also determined. Cell proliferation assays were performed in the presence of VEGF-A165 plus the 59.8-kDa VEGFR1 fragment or VEGF-A165 alone. RESULTS: Matrix metalloproteinase 14 binds and cleaves rmVEGFR1 to produce 59.8-kDa (N-terminal fragment, Ig domains 1-5), 35-kDa (C-terminal fragment containing IgG and His-tag), and 21-kDa (Ig domains 6-7) fragments. The 59.8-kDa fragment showed binding to VEGF-A165 and inhibited VEGF-induced endothelial cell mitogenesis. CONCLUSIONS: Our findings suggest that VEGFR1 cleavage by MMP14 in the cornea leads to a VEGF-trap effect, reducing the proangiogenic effect of VEGF-A165, thereby reducing corneal angiogenesis.


Asunto(s)
Córnea/patología , Neovascularización de la Córnea/prevención & control , Metaloproteinasa 14 de la Matriz/metabolismo , Factor A de Crecimiento Endotelial Vascular/efectos de los fármacos , Receptor 1 de Factores de Crecimiento Endotelial Vascular/efectos de los fármacos , Animales , Western Blotting , Movimiento Celular , Proliferación Celular/efectos de los fármacos , Córnea/efectos de los fármacos , Córnea/metabolismo , Neovascularización de la Córnea/metabolismo , Neovascularización de la Córnea/patología , Modelos Animales de Enfermedad , Electroforesis en Gel de Poliacrilamida , Ratones , Factor A de Crecimiento Endotelial Vascular/metabolismo , Receptor 1 de Factores de Crecimiento Endotelial Vascular/metabolismo
13.
Zhonghua Wei Zhong Bing Ji Jiu Yi Xue ; 27(2): 127-32, 2015 Feb.
Artículo en Chino | MEDLINE | ID: mdl-25665612

RESUMEN

OBJECTIVE: To investigate the effects of Xuebijing injection on hemodynamics, cardiac function, and endothelial function in patients with severe sepsis in order to study the therapeutic effect of Xuebijing in the treatment of severe sepsis. METHODS: A prospective randomized controlled trial was conducted. Sixty-six severe sepsis patients admitted to the Department of Critical Care Medicine of Guangdong Hospital of Traditional Chinese Medicine from March 2013 to February 2014 were enrolled. The patients were divided into control group (n = 31) and Xuebijing group (n = 35). The patients in both groups were treated according to "2012 international guidelines for management of severe sepsis and septic shock", and the patients in Xuebijing group received Xuebijing injection of 50 mL(added with 100 mL of 0.9% sodium chloride injection) twice a day for 5 days, and those in control group received instead 150 mL of 0.9% sodium chloride injection for 5 days. The heart rate (HR), mean arterial pressure (MAP), central venous pressure (CVP), and dosage of vasoactive drugs before and 1 day and 5 days after treatment were determined for hemodynamics assessment. Blood lactic acid (Lac), central venous oxygen saturation (ScvO2), and difference in arterial-venous blood carbon dioxide pressure (Pv-aCO2) were determined for microcirculation assessment. The left ventricular ejection fraction (LVEF), cardiac output (CO), left ventricular end diastolic diameter (LVEDD), the ratio of blood flow of mitral orifice between rapid filling period and atrial systole period (E/A), and B-type natriuretic peptide (BNP) were determined for cardiac function assessment. Vascular endothelial growth factor (VEGF) and soluble receptor (sFLT-1) were assessed for endothelial function assessment. The relationship among the indexes of the hemodynamics, microcirculation, cardiac function, and endothelial function was analyzed with Pearson related-analysis. RESULTS: After treatment, HR, MAP, CVP, Lac, ScvO2, and Pv-aCO2 were improved in both groups compared with those before treatment, and the dosage of norepinephrine (NE) was decreased in Xuebijing group. Compared with control group, MAP at 5 days after treatment in Xuebijing group was significantly increased [mmHg (1 mmHg = 0.133 kPa): 74.9±10.7 vs. 70.2±6.6, P < 0.05], the dosage of NE was decreased [µg×kg-1×min-1: 0.01 (0.00, 0.22) vs. 0.10 (0.05, 0.80), P < 0.05], LVEF was significantly increased (0.617±0.125 vs. 0.533±0.129, P < 0.05), BNP was significantly decreased [ng/L: 117.3 (52.0, 443.0) vs. 277.2 (67.9, 2 370.2), P < 0.05], while VEGF showed no significant change (ng/L: 101.1±23.2 vs. 89.6±20.5, P > 0.05), and sFLT-1 was significantly decreased (ng/L: 245.7±86.2 vs. 295.1±95.1, P < 0.05). It was shown by Pearson coefficient bivariate correlation analysis that sFLT-1 was negatively correlated with MAP and ScvO2 (r = -0.569, P = 0.000; r = -0.341, P = 0.008) 5 days after treatment, while it was positively associated with Lac and acute physiology and chronic health evaluation II (APACHE II) score (r = 0.749, P = 0.000; r = 0.645, P = 0.000). CONCLUSIONS: After treatment, HR, MAP, CVP, Lac, ScvO2, and Pv-aCO2 were improved in both groups compared with those before treatment, and the dosage of norepinephrine (NE) was decreased in Xuebijing group. Compared with control group, MAP at 5 days after treatment in Xuebijing group was significantly increased [mmHg (1 mmHg = 0.133 kPa): 74.9±10.7 vs. 70.2±6.6, P < 0.05], the dosage of NE was decreased [µg×kg-1×min-1: 0.01 (0.00, 0.22) vs. 0.10 (0.05, 0.80), P < 0.05], LVEF was significantly increased (0.617±0.125 vs. 0.533±0.129, P < 0.05), BNP was significantly decreased [ng/L: 117.3 (52.0, 443.0) vs. 277.2 (67.9, 2 370.2), P < 0.05], while VEGF showed no significant change (ng/L: 101.1±23.2 vs. 89.6±20.5, P > 0.05), and sFLT-1 was significantly decreased (ng/L: 245.7±86.2 vs. 295.1±95.1, P < 0.05). It was shown by Pearson coefficient bivariate correlation analysis that sFLT-1 was negatively correlated with MAP and ScvO2 (r = -0.569, P = 0.000; r = -0.341, P = 0.008) 5 days after treatment, while it was positively associated with Lac and acute physiology and chronic health evaluation II (APACHE II) score (r = 0.749, P = 0.000; r = 0.645, P = 0.000).


Asunto(s)
Medicamentos Herbarios Chinos/farmacología , Endotelio/efectos de los fármacos , Hemodinámica/efectos de los fármacos , Sepsis/tratamiento farmacológico , APACHE , Dióxido de Carbono/sangre , Gasto Cardíaco , Presión Venosa Central , Endotelio/metabolismo , Humanos , Microcirculación , Péptido Natriurético Encefálico , Norepinefrina , Oximetría , Estudios Prospectivos , Choque Séptico , Factor A de Crecimiento Endotelial Vascular/efectos de los fármacos , Receptor 1 de Factores de Crecimiento Endotelial Vascular/efectos de los fármacos
14.
Hypertension ; 65(4): 855-62, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25667213

RESUMEN

Preeclampsia is a severe complication of pregnancy, characterized by hypertension, oxidative stress, and severe endothelial dysfunction. Antiangiogenic factors, soluble fms-like tyrosine kinase-1 (sFlt-1) and soluble endoglin, play key pathophysiological roles in preeclampsia. Heme oxygenase-1 (HO-1) is a cytoprotective, antioxidant enzyme reported to be downregulated in preeclampsia. Studies propose that inducing HO-1 may also decrease sFlt-1 production. Sofalcone, a gastric antiulcer agent in clinical use, is known to induce HO-1 in gastric epithelium. We aimed to investigate whether sofalcone induces HO-1 and reduces sFlt-1 release from primary human placental and endothelial cells and blocks endothelial dysfunction in vitro. We isolated human trophoblasts and endothelial cells (human umbilical vein endothelial cells) and also used uterine microvascular cells. We investigated the effects of sofalcone on (1) HO-1 production, (2) activation of the nuclear factor (erythroid-derived 2)-like 2 pathway, (3) sFlt-1 and soluble endoglin release, (4) tumor necrosis factor α-induced monocyte adhesion and vascular cell adhesion molecule upregulation, and (5) endothelial tubule formation. Sofalcone potently increased HO-1 mRNA and protein in both primary trophoblasts and human umbilical vein endothelial cells. Furthermore, sofalcone treatment caused nuclear translocation of nuclear factor (erythroid-derived 2)-like 2 and transactivation of other nuclear factor (erythroid-derived 2)-like 2 responsive genes (NQO1, TXN, and GCLC). Importantly, sofalcone significantly decreased the secretion of sFlt-1 from primary human trophoblasts. Sofalcone potently suppressed endothelial dysfunction in 2 in vitro models, blocking tumor necrosis factor α-induced monocyte adhesion and vascular cell adhesion molecule 1 expression in human umbilical vein endothelial cells. These results indicate that in primary human tissues, sofalcone can potently activate antioxidant nuclear factor (erythroid-derived 2)-like 2/HO-1 pathway, decrease sFlt-1 production, and ameliorate endothelial dysfunction. We propose that sofalcone is a novel therapeutic candidate for preeclampsia.


Asunto(s)
Chalconas/farmacología , Hemo-Oxigenasa 1/metabolismo , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Factor 2 Relacionado con NF-E2/genética , Preeclampsia/genética , Regulación hacia Arriba/genética , Receptor 1 de Factores de Crecimiento Endotelial Vascular/metabolismo , Western Blotting , Supervivencia Celular , Células Cultivadas , Ensayo de Inmunoadsorción Enzimática , Femenino , Células Endoteliales de la Vena Umbilical Humana/patología , Humanos , Factor 2 Relacionado con NF-E2/biosíntesis , Factor 2 Relacionado con NF-E2/efectos de los fármacos , Estrés Oxidativo , Preeclampsia/metabolismo , Preeclampsia/terapia , Embarazo , ARN/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal/genética , Activación Transcripcional , Trofoblastos/metabolismo , Trofoblastos/patología , Receptor 1 de Factores de Crecimiento Endotelial Vascular/efectos de los fármacos
15.
Gynecol Obstet Invest ; 79(2): 90-6, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25633310

RESUMEN

BACKGROUND: Preeclampsia (PE) is associated with oxidative stress in the maternal circulation and placenta. This study aimed to determine if inhibition of lectin-like oxidized low-density lipoprotein receptor 1 (LOX-1) gives protection against oxidative stress-mediated trophoblast dysfunction. METHODS: Plasma and placenta samples were obtained from 106 women with PE and 106 women with normal pregnancy (NP). Oxidized low-density lipoprotein (oxLDL) and soluble LOX-1 levels were determined by enzyme-linked immunoassay. Placental LOX-1 expression was determined by western blotting. Trophoblasts were subjected to hypoxia and treated with pooled plasma from patients with PE. Expression levels of placenta growth factor (PIGF) and the soluble form of the PIGF receptor (sFlt-1) in trophoblasts were determined. RESULTS: Plasma concentrations of oxLDL and sLOX-1 were significantly over-expressed and LOX-1 protein expression in the placenta was significantly increased in PE patients compared with matched NP controls (both p < 0.05). Exposure of trophoblasts to hypoxia and pooled PE plasma induced overexpression of sFlt-1 and downregulation of PIGF. These effects were inhibited by the LOX-1 inhibitor TS20. CONCLUSION: LOX-1 accumulation may contribute to the pathogenesis of PE by promoting sFlt-1 production in trophoblasts, suggesting that oxidative stress may be an important mediator regulating angiogenic pathways in trophoblasts.


Asunto(s)
Lipoproteínas LDL/metabolismo , Estrés Oxidativo/fisiología , Placenta/metabolismo , Preeclampsia/metabolismo , Proteínas Gestacionales/metabolismo , Receptores Depuradores de Clase E/metabolismo , Receptor 1 de Factores de Crecimiento Endotelial Vascular/metabolismo , Adulto , Femenino , Humanos , Lipoproteínas LDL/sangre , Factor de Crecimiento Placentario , Preeclampsia/sangre , Embarazo , Proteínas Gestacionales/efectos de los fármacos , Receptores Depuradores de Clase E/antagonistas & inhibidores , Receptores Depuradores de Clase E/sangre , Trofoblastos/metabolismo , Trofoblastos/patología , Receptor 1 de Factores de Crecimiento Endotelial Vascular/efectos de los fármacos
16.
J Periodontol ; 86(1): 162-72, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25327303

RESUMEN

BACKGROUND: Vascular endothelial growth factor (VEGF) plays an important role during angiogenesis and bone repair. This study investigated whether the use of meloxicam alters bone repair via downregulation of VEGF and receptor expression. METHODS: One hundred twenty male Wistar rats had their maxillary right incisor extracted. Animals were divided into a control group (CG; n = 60) and a meloxicam-treated group (TG; n = 60) that received either a single daily intraperitoneal injection of 0.9% NaCl or meloxicam 3 mg/kg, respectively, for 7 consecutive days. Alveolar bone repair was evaluated histomorphometrically, whereas VEGF and its receptors were analyzed by immunohistochemistry and quantitative polymerase chain reaction (qPCR). Data were submitted to two-way analysis of variance and Tukey post hoc test with P < 0.05. RESULTS: Bone volume density increased significantly (P = 0.001) in both groups with a strong correlation between treatment and periods (P = 0.003). In the TG, a small amount of bone formation occurred compared with the CG between 3 and 21 days. No significant differences in the number of VEGF-positive cells per square millimeter (P = 0.07) and VEGF messenger RNA (mRNA) expression (P = 0.49) were found between groups. Immunostained cells per square millimeter and mRNA expression for VEGF receptor (VEGFR)-1 (P = 0.04 and P < 0.001) and VEGFR-2 (P < 0.001 for both analysis) showed a strong interaction between treatment groups and periods. In the TG, immunostained cells per square millimeter and mRNA expression for VEGFR-1 were, respectively, 89% and 37% lower from 3 to 10 days compared with the CG, whereas for VEGFR-2, these values were 252% and 60%, respectively, from 3 to 7 days. CONCLUSION: In rat alveolar bone repair, meloxicam did not affect VEGF expression but downregulated VEGFR expression, which may cause a delay in the bone repair/remodeling process.


Asunto(s)
Proceso Alveolar/efectos de los fármacos , Antiinflamatorios no Esteroideos/farmacología , Remodelación Ósea/efectos de los fármacos , Inhibidores de la Ciclooxigenasa 2/farmacología , Isoenzimas/antagonistas & inhibidores , Tiazinas/farmacología , Tiazoles/farmacología , Receptor 1 de Factores de Crecimiento Endotelial Vascular/efectos de los fármacos , Receptor 2 de Factores de Crecimiento Endotelial Vascular/efectos de los fármacos , Animales , Densidad Ósea/efectos de los fármacos , Regulación hacia Abajo , Masculino , Maxilar/efectos de los fármacos , Meloxicam , Osteoclastos/patología , Osteogénesis/efectos de los fármacos , Ratas , Ratas Wistar , Transducción de Señal/efectos de los fármacos , Factores de Tiempo , Alveolo Dental/efectos de los fármacos , Factor A de Crecimiento Endotelial Vascular/efectos de los fármacos
17.
Oncol Rep ; 32(4): 1385-94, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25109488

RESUMEN

The effects of mead acid (MA; 5,8,11-eicosatrienoic acid) on the suppression of breast cancer cell growth and metastasis were examined in vitro and in vivo by using the KPL-1 human breast cancer cell line. MA suppressed KPL-1 cell growth in culture with an IC50 value of 214.2 µM (65.7 µg/ml) for 72 h, and MA significantly suppressed transplanted KPL-1 tumor growth (tumor volume and tumor weight: 872±103 mm3 and 1,000±116 mg vs. 376±66 mm3 and 517±84 mg) and regional (axillary) lymph node metastasis (67%, 10/15 vs. 10%, 1/10) in female athymic mice fed an MA-rich diet for 8 weeks. Tumor suppression was due to the suppression of cell proliferation. In ELISA, although vascular endothelial growth factor (VEGF) levels were unchanged, VEGF receptor (VEGFR)1 and VEGFR2 levels were significantly decreased after treatment with a 214.2-µM dose of MA for 72 h; E-cadherin levels were unchanged. As VEGF, VEGFR1 and VEGFR2 expression was co-localized in KPL-1 cells, the mechanism leading to cell growth suppression was VEGF signaling directly to KPL-1 cells by an autocrine process. In contrast, MA did not influence angiogenesis. The mechanisms of action were through VEGF signaling directly to cancer cells.


Asunto(s)
Ácido 8,11,14-Eicosatrienoico/análogos & derivados , Apoptosis/efectos de los fármacos , Neoplasias de la Mama/metabolismo , Proliferación Celular/efectos de los fármacos , Ácido 8,11,14-Eicosatrienoico/farmacología , Animales , Cadherinas/efectos de los fármacos , Cadherinas/metabolismo , Línea Celular Tumoral , Ensayos de Selección de Medicamentos Antitumorales , Femenino , Humanos , Técnicas In Vitro , Ratones , Ratones Endogámicos BALB C , Trasplante de Neoplasias , Neovascularización Patológica , Factor A de Crecimiento Endotelial Vascular/efectos de los fármacos , Factor A de Crecimiento Endotelial Vascular/metabolismo , Receptor 1 de Factores de Crecimiento Endotelial Vascular/efectos de los fármacos , Receptor 1 de Factores de Crecimiento Endotelial Vascular/metabolismo , Receptor 2 de Factores de Crecimiento Endotelial Vascular/efectos de los fármacos , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
18.
Am J Obstet Gynecol ; 211(5): 541.e1-8, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-24793974

RESUMEN

OBJECTIVE: Diabetes mellitus is a risk factor for preeclampsia. Cytotrophoblast (CTB) invasion is facilitated from the conversion of plasminogen to plasmin by urokinase plasminogen activator (uPA), regulated by plasminogen activator inhibitor 1 (PAI-1), and may be inhibited in preeclampsia. This study assessed signaling mechanisms of hyperglycemia-induced CTB dysfunction. STUDY DESIGN: Human CTBs were treated with 45, 135, 225, 495, or 945 mg/dL glucose for 48 hours. Some cells were pretreated with a p38 inhibitor (SB203580) or a peroxisome proliferator-activated receptor-gamma (PPAR-γ) ligand (rosiglitazone). Expression of uPA, PAI-1, and PPAR-γ levels and p38 mitogen-activated protein kinase phosphorylation were measured by Western blot in cell lysates. Messenger ribonucleic acid of uPA and PAI-1 was measured by quantitative polymerase chain reaction. Levels of interleukin-6, angiogenic (vascular endothelial growth factor [VEGF], placenta growth factor [PlGF]) and antiangiogenic factors (soluble fms-like tyrosine kinase-1 [sFlt-1], soluble endoglin [sEng]) were measured in the media by enzyme-linked immunosorbent assay kits. Statistical comparisons were performed using analysis of variance with a Duncan's post-hoc test. RESULTS: Both uPA and PAI-1 protein and messenger ribonucleic acid were down-regulated (P < .05) in CTBs treated with 135 mg/dL glucose or greater compared with basal (45 mg/dL). The sEng, sFlt-1, and interleukin-6 were up-regulated, whereas the VEGF and PlGF were down-regulated by 135 mg/dL glucose or greater. p38 phosphorylation and PPAR-γ were up-regulated (P < .05) in hyperglycemia-treated CTBs. The SB203580 or rosiglitazone pretreatment showed an attenuation of glucose-induced down-regulation of uPA and PAI-1. CONCLUSION: Hyperglycemia disrupts the invasive profile of CTB by decreasing uPA and PAI-1 expression; down-regulating VEGF and PlGF; and up-regulating sEng, sFlt-1, and interleukin-6. Attenuation of CTB dysfunction by SB203580 or rosiglitazone pretreatment suggests the involvement of stress signaling.


Asunto(s)
Glucosa/farmacología , Hiperglucemia/metabolismo , Inhibidor 1 de Activador Plasminogénico/genética , Trofoblastos/efectos de los fármacos , Activador de Plasminógeno de Tipo Uroquinasa/efectos de los fármacos , Antígenos CD/efectos de los fármacos , Antígenos CD/metabolismo , Diabetes Gestacional/metabolismo , Endoglina , Ensayo de Inmunoadsorción Enzimática , Femenino , Humanos , Hipoglucemiantes/farmacología , Imidazoles/farmacología , Interleucina-6/metabolismo , PPAR gamma/efectos de los fármacos , PPAR gamma/metabolismo , Fosforilación/efectos de los fármacos , Factor de Crecimiento Placentario , Inhibidor 1 de Activador Plasminogénico/metabolismo , Reacción en Cadena de la Polimerasa , Embarazo , Proteínas Gestacionales/efectos de los fármacos , Proteínas Gestacionales/metabolismo , Piridinas/farmacología , Receptores de Superficie Celular/efectos de los fármacos , Receptores de Superficie Celular/metabolismo , Rosiglitazona , Transducción de Señal/efectos de los fármacos , Tiazolidinedionas/farmacología , Activador de Plasminógeno de Tipo Uroquinasa/genética , Activador de Plasminógeno de Tipo Uroquinasa/metabolismo , Factor A de Crecimiento Endotelial Vascular/efectos de los fármacos , Factor A de Crecimiento Endotelial Vascular/metabolismo , Receptor 1 de Factores de Crecimiento Endotelial Vascular/efectos de los fármacos , Receptor 1 de Factores de Crecimiento Endotelial Vascular/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/antagonistas & inhibidores , Proteínas Quinasas p38 Activadas por Mitógenos/efectos de los fármacos , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
20.
Nephrol Dial Transplant ; 29(6): 1225-31, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24516233

RESUMEN

BACKGROUND: Soluble Flt1 (sFlt1) is a potent inhibitor of vascular endothelial growth factor, secreted mainly by the placenta, endothelial cells and monocytes. Increased sFlt1 serum levels correlate with endothelial dysfunction and cardiovascular complications in dialysis patients. However, the impact of dialysis by itself on sFlt1 serum levels remains unknown. METHODS: We assessed sFlt1 kinetics during dialysis and the impact of different dialysis techniques [high-flux haemodialysis (HD), haemodiafiltration (HDF)] and heparinization procedures on sFlt1 serum levels in 48 patients on regular dialysis. RESULTS: sFlt1 serum levels increased as early as 1 min after the start of dialysis and peaked at 15 min before returning to baseline at 4 h [mean peak level 2551 pg/mL, versus 102 before dialysis (P < 0.0001)]. sFlt1 kinetics were similar with two different dialysis membranes. In contrast, when unfractionated heparin (UH) and low-molecular-weight heparin (LMWH) were omitted during dialysis (HD or pre-dilution HDF), no significant increase in sFlt1 levels occurred. Conversely, delayed administration of LMWH (after 30 min of a heparin-free HD) induced a sharp increase in sFlt1. Similarly, when UH and LMWH were omitted and citrate-based dialysate or a heparin-coated membrane was used, sFlt1 levels remained unchanged. When heparinization procedures were the same, no difference in sFlt1 levels was noted between HD and HDF. In vitro, UH and LMWH failed to induce sFlt1 release by monocytes from controls or HD patients. These findings suggest that priming of monocytes on the extracorporeal circuit is required for heparin-induced sFlt1 release or that endothelial cells contribute to this increase. CONCLUSIONS: Our results indicate that heparin-based HD induces a major sFlt1 release, which may exacerbate the anti-angiogenic state and thus endothelial dysfunction, commonly found in dialysis patients.


Asunto(s)
Anticoagulantes/farmacología , Heparina/farmacología , Fallo Renal Crónico/sangre , Fallo Renal Crónico/terapia , Neovascularización Fisiológica/efectos de los fármacos , Diálisis Renal , Receptor 1 de Factores de Crecimiento Endotelial Vascular/sangre , Receptor 1 de Factores de Crecimiento Endotelial Vascular/efectos de los fármacos , Adulto , Anciano , Anciano de 80 o más Años , Células Endoteliales/metabolismo , Endotelio Vascular/citología , Endotelio Vascular/fisiopatología , Femenino , Hemodiafiltración , Heparina de Bajo-Peso-Molecular/farmacología , Humanos , Masculino , Membranas Artificiales , Persona de Mediana Edad , Embarazo , Diálisis Renal/métodos , Adulto Joven
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...