Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
1.
Thromb Haemost ; 121(12): 1628-1636, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-33851387

RESUMEN

BACKGROUND: Platelet cytosolic cyclic adenosine monophosphate (cAMP) levels are balanced by synthesis, degradation, and efflux. Efflux can occur via multidrug resistant protein-4 (MRP4; ABCC4) present on dense granule and/or plasma membranes. As lipid rafts have been shown to interfere on cAMP homeostasis, we evaluated the relationships between the distribution and activity of MRP4 in lipid rafts and cAMP efflux. METHODS: Platelet activation and cAMP homeostasis were analyzed in human and wild-type or MRP4-deleted mouse platelets in the presence of methyl-ß-cyclodextrin (MßCD) to disrupt lipid rafts, and of activators of the cAMP signalling pathways. Human platelet MRP4 and effector proteins of the cAMP pathway were analyzed by immunoblots in lipid rafts isolated by differential centrifugation. RESULTS: MßCD dose dependently inhibited human and mouse platelet aggregation without affecting per se cAMP levels. An additive inhibitory effect existed between the adenylate cyclase (AC) activator forskolin and MßCD that was accompanied by an overincrease of cAMP, and which was significantly enhanced upon MRP4 deletion. Finally, an efflux of cAMP out of resting platelets incubated with prostaglandin E1 (PGE1) was observed that was partly dependent on MRP4. Lipid rafts contained a small fraction (≈15%) of MRP4 and most of the inhibitory G-protein Gi, whereas Gs protein, AC3, and phosphodiesterases PDE2 and PDE3A were all present as only trace amounts. CONCLUSION: Our results are in favour of part of MRP4 present at the platelet surface, including in lipid rafts. Lipid raft integrity is necessary for cAMP signalling regulation, although MRP4 and most players of cAMP homeostasis are essentially located outside rafts.


Asunto(s)
Plaquetas/metabolismo , AMP Cíclico/sangre , Microdominios de Membrana/metabolismo , Proteínas Asociadas a Resistencia a Múltiples Medicamentos/sangre , Agregación Plaquetaria , Sistemas de Mensajero Secundario , Alprostadil/farmacología , Animales , Plaquetas/efectos de los fármacos , Microdominios de Membrana/efectos de los fármacos , Microdominios de Membrana/genética , Ratones Noqueados , Proteínas Asociadas a Resistencia a Múltiples Medicamentos/genética , Agregación Plaquetaria/efectos de los fármacos , beta-Ciclodextrinas/farmacología
2.
Angiogenesis ; 23(2): 249-264, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-31900750

RESUMEN

INTRODUCTION: Although thioredoxin-interacting protein (TXNIP) is involved in a variety of biological functions, the contribution of endothelial TXNIP has not been well-defined in regards to endothelial and vascular function or in post-ischemic revascularisation. We postulated that inhibition of endothelial TXNIP with siRNA or in a Cre-LoxP system could be involved in protection from high fat, high protein, low carbohydrate (HFHPLC) diet-induced oxidative stress and endothelial dysfunction, leading to vascular damage and impaired revascularisation in vivo. METHODS AND RESULTS: To investigate the role of endothelial TXNIP, the TXNIP gene was deleted in endothelial cells using anti-TXNIP siRNA treatment or the Cre-LoxP system. Murine models were fed a HFHPLC diet, known to induce metabolic disorders. Endothelial TXNIP targeting resulted in protection against metabolic disorder-related endothelial oxidative stress and endothelial dysfunction. This protective effect mitigates media cell loss induced by metabolic disorders and hampered metabolic disorder-related vascular dysfunction assessed by aortic reactivity and distensibility. In aortic ring cultures, metabolic disorders impaired vessel sprouting and this alteration was alleviated by deletion of endothelial TXNIP. When subjected to ischemia, mice fed a HFHPLC diet exhibited defective post-ischemic angiogenesis and impaired blood flow recovery in hind limb ischemia. However, reducing endothelial TXNIP rescued metabolic disorder-related impairment of ischemia-induced revascularisation. CONCLUSION: Collectively, these results show that targeting endothelial TXNIP in metabolic disorders is essential to maintaining endothelial function, vascular function and improving ischemia-induced revascularisation, making TXNIP a potential therapeutic target for therapy of vascular complications related to metabolic disorders.


Asunto(s)
Proteínas Portadoras/genética , Células Endoteliales/fisiología , Isquemia , Enfermedades Metabólicas/fisiopatología , Neovascularización Fisiológica/genética , Tiorredoxinas/genética , Animales , Células Cultivadas , Citoprotección/genética , Miembro Posterior/irrigación sanguínea , Isquemia/genética , Isquemia/metabolismo , Isquemia/fisiopatología , Isquemia/prevención & control , Masculino , Enfermedades Metabólicas/complicaciones , Enfermedades Metabólicas/genética , Enfermedades Metabólicas/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Estrés Oxidativo/fisiología
3.
Arterioscler Thromb Vasc Biol ; 39(10): e219-e232, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31434496

RESUMEN

OBJECTIVE: Ceramide 1-phosphate (C1P) is a bioactive sphingolipid highly augmented in damaged tissues. Because of its abilities to stimulate migration of murine bone marrow-derived progenitor cells, it has been suggested that C1P might be involved in tissue regeneration. In the present study, we aimed to investigate whether C1P regulates survival and angiogenic activity of human progenitor cells with great therapeutic potential in regenerative medicine such as endothelial colony-orming cells (ECFCs). Approach and Results: C1P protected ECFC from TNFα (tumor necrosis factor-α)-induced and monosodium urate crystal-induced death and acted as a potent chemoattractant factor through the activation of ERK1/2 (extracellular signal-regulated kinases 1 and 2) and AKT pathways. C1P treatment enhanced ECFC adhesion to collagen type I, an effect that was prevented by ß1 integrin blockade, and to mature endothelial cells, which was mediated by the E-selectin/CD44 axis. ECFC proliferation and cord-like structure formation were also increased by C1P, as well as vascularization of gel plug implants loaded or not with ECFC. In a murine model of hindlimb ischemia, local administration of C1P alone promoted blood perfusion and reduced necrosis in the ischemic muscle. Additionally, the beneficial effects of ECFC infusion after ischemia were amplified by C1P pretreatment, resulting in a further and significant enhancement of leg reperfusion and muscle repair. CONCLUSIONS: Our findings suggest that C1P may have therapeutic relevance in ischemic disorders, improving tissue repair by itself, or priming ECFC angiogenic responses such as chemotaxis, adhesion, proliferation, and tubule formation, which result in a better outcome of ECFC-based therapy.


Asunto(s)
Apoptosis/efectos de los fármacos , Ceramidas/farmacología , Células Progenitoras Endoteliales/metabolismo , Neovascularización Fisiológica/efectos de los fármacos , Regeneración/efectos de los fármacos , Animales , Diferenciación Celular , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Modelos Animales de Enfermedad , Células Progenitoras Endoteliales/efectos de los fármacos , Humanos , Isquemia/tratamiento farmacológico , Isquemia/metabolismo , Ratones , Morfogénesis/efectos de los fármacos , Sensibilidad y Especificidad
4.
Artículo en Inglés | MEDLINE | ID: mdl-31442897

RESUMEN

Discrepancies in preclinical studies of aspirin (ASA) antiplatelet activity in mouse models of bleeding and arterial thrombosis led us to evaluate commonly reported methods in order to propose a procedure for reliably measuring the effects of single dose ASA on mouse hemostasis. FVB and C57Bl6 mice received 100 mg/kg of ASA or vehicle orally 30 min or 3 h prior to investigate either hemostasis using the tail bleeding assay or carotid thrombosis induced by FeCl3, or to blood sampling for isolated platelet aggregation and TXB2 generation. Expected inhibition of COX1 by ASA was ascertained by a strong decrease in TXB2 production, and its effect on platelet function and hemostasis, by decreased collagen-induced aggregation and increased bleeding time, respectively. Strikingly, we determined that anti-hemostatic effects of ASA were more predictable 30 min after administration than 3 h later. Conversely, ASA did not alter time to arterial occlusion of the carotid upon FeCl3-induced thrombosis, suggesting ASA not to be used as reference inhibitor drug in this model of arterial thrombosis.


Asunto(s)
Aspirina/administración & dosificación , Aspirina/farmacología , Evaluación Preclínica de Medicamentos/métodos , Hemostasis/efectos de los fármacos , Inhibidores de Agregación Plaquetaria/administración & dosificación , Inhibidores de Agregación Plaquetaria/farmacología , Animales , Aspirina/uso terapéutico , Modelos Animales de Enfermedad , Hemorragia/tratamiento farmacológico , Masculino , Ratones , Ratones Endogámicos C57BL , Agregación Plaquetaria/efectos de los fármacos , Inhibidores de Agregación Plaquetaria/uso terapéutico , Pruebas de Función Plaquetaria , Trombosis/tratamiento farmacológico
5.
Stem Cell Rev Rep ; 15(4): 590-600, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-30879244

RESUMEN

Stem cells at the origin of endothelial progenitor cells and in particular endothelial colony forming cells (ECFCs) subtype have been largely supposed to be positive for the CD133 antigen, even though no clear correlation has been established between its expression and function in ECFCs. We postulated that CD133 in ECFCs might be expressed intracellularly, and could participate to vasculogenic properties. ECFCs extracted from cord blood were used either fresh (n = 4) or frozen (n = 4), at culture days <30, to investigate the intracellular presence of CD133 by flow cytometry and confocal analysis. Comparison with HUVEC and HAEC mature endothelial cells was carried out. Then, CD133 was silenced in ECFCs using specific siRNA (siCD133-ECFCs) or scramble siRNA (siCtrl-ECFCs). siCD133-ECFCs (n = 12), siCtrl-ECFCs (n = 12) or PBS (n = 12) were injected in a hind-limb ischemia nude mouse model and vascularization was quantified at day 14 with H&E staining and immunohistochemistry for CD31. Results of flow cytometry and confocal microscopy evidenced the positivity of CD133 in ECFCs after permeabilization compared with not permeabilized ECFCs (p < 0.001) and mature endothelial cells (p < 0.03). In the model of mouse hind-limb ischemia, silencing of CD133 in ECFCs significantly abolished post-ischemic revascularization induced by siCtrl-ECFCs; indeed, a significant reduction in cutaneous blood flows (p = 0.03), capillary density (CD31) (p = 0.01) and myofiber regeneration (p = 0.04) was observed. Also, a significant necrosis (p = 0.02) was observed in mice receiving siCD133-ECFCs compared to those treated with siCtrl-ECFCs. In conclusion, our work describes for the first time the intracellular expression of the stemness marker CD133 in ECFCs. This feature could resume the discrepancies found in the literature concerning CD133 positivity and ontogeny in endothelial progenitors.


Asunto(s)
Antígeno AC133/biosíntesis , Antígenos de Diferenciación/biosíntesis , Células Progenitoras Endoteliales/metabolismo , Regulación de la Expresión Génica , Neovascularización Fisiológica , Animales , Células Progenitoras Endoteliales/citología , Xenoinjertos , Miembro Posterior/irrigación sanguínea , Células Endoteliales de la Vena Umbilical Humana/citología , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Humanos , Isquemia/metabolismo , Isquemia/patología , Isquemia/terapia , Masculino , Ratones , Ratones Desnudos , Trasplante de Células Madre
6.
Ultraschall Med ; 40(6): 734-742, 2019 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-30241104

RESUMEN

OBJECTIVE: Vascular Ehlers-Danlos syndrome (vEDS) is associated with arterial ruptures due to a mutant gene encoding collagen type III (Col-III). To better understand the role of Col-III, we aimed at evaluating aortic stiffness and dynamic stiffening in vEDS mouse models, with either a quantitative (col3KO mice) or a qualitative Col-III defect (col3KI mice). MATERIALS AND METHODS: Abdominal aortic wall pulse wave velocities (PWV) in col3KO and col3KI mice were compared to their respective wild type (WT) littermates using a 15 MHz ultrafast ultrasonic transducer. A carotid catheter continuously monitored pressure changes due to phenylephrine injections. PWV1, generated at diastolic blood pressure (DBP), and PWV2, at systolic blood pressure (SBP) were recorded. Difference between PWV2 and PWV1 (Delta-PWV) normalized by the pulse pressure (PP), corresponding to the aortic stiffening over the cardiac cycle, were compared between mutant and WT mice, as well as the regression slope of PWV as a function of pressure. RESULTS: Delta-PWV/PP was lower in col3KO (p = 0.033) and col3KI mice (p < 0.001) vs. WT-mice regardless of the pressure level. The slope of PWV1 with DBP increase showed a lower arterial stiffness in mutant mice vs. controls in both models. This difference was amplified when evaluating stiffness at systolic blood pressure levels with PWV2. CONCLUSION: In both vEDS mouse models, aortic stiffening was reduced, mainly driven by a lower stiffness at systolic blood pressure. Defective Col-III may be responsible for this, as it is utilized when pressure rises. These pre-clinical data could explain vascular fragility observed in vEDS patients.


Asunto(s)
Síndrome de Ehlers-Danlos , Hipertensión , Rigidez Vascular , Animales , Presión Sanguínea , Síndrome de Ehlers-Danlos/diagnóstico por imagen , Humanos , Ratones , Fenotipo , Análisis de la Onda del Pulso , Ultrasonografía
7.
Stem Cell Rev Rep ; 14(6): 812-822, 2018 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-30267203

RESUMEN

Idiopathic pulmonary fibrosis (IPF) is a devastating lung disease characterized by fibroblast proliferation, extracellular matrix deposition, destruction of pulmonary alveolar architecture and vascular remodeling. Apart pirfenidone or nintendanib that only slow down the fibrotic process, there is no curative treatment other than lung transplantation. Because cell therapy approaches have been proposed in IPF, we hypothesized that injection of endothelial colony-forming cells (ECFCs), the vasculogenic subtype of endothelial progenitor cells, could modulate fibrosis in a Nude mouse model of bleomycin induced-pulmonary fibrosis. Mice were injected with ECFCs isolated from cord blood and from peripheral blood of adult IPF patients at two time-points: during the development of the fibrosis or once the fibrosis was constituted. We assessed morbidity, weight variation, collagen deposition, lung imaging by microCT, Fulton score and microvascular density. Neither ECFCs isolated from cord blood nor from IPF patients were able to modulate fibrosis or vascular density during fibrogenesis or when fibrosis was constituted. These findings indicate that human ECFCs do not promote an adaptive regenerative response in the lung upon fibrosis or angiogenic process in the setting of bleomycin-induced pulmonary fibrosis in Nude mice.


Asunto(s)
Bleomicina/efectos adversos , Células Progenitoras Endoteliales/metabolismo , Pulmón , Fibrosis Pulmonar , Microtomografía por Rayos X , Animales , Bleomicina/farmacología , Modelos Animales de Enfermedad , Células Progenitoras Endoteliales/patología , Humanos , Pulmón/diagnóstico por imagen , Pulmón/metabolismo , Ratones , Ratones Desnudos , Fibrosis Pulmonar/inducido químicamente , Fibrosis Pulmonar/diagnóstico por imagen , Fibrosis Pulmonar/metabolismo
8.
Stem Cell Res Ther ; 9(1): 120, 2018 05 02.
Artículo en Inglés | MEDLINE | ID: mdl-29720269

RESUMEN

BACKGROUND: We have previously demonstrated that acidic preconditioning of human endothelial colony-forming cells (ECFC) increased proliferation, migration, and tubulogenesis in vitro, and increased their regenerative potential in a murine model of hind limb ischemia without baseline disease. We now analyze whether this strategy is also effective under adverse conditions for vasculogenesis, such as the presence of ischemia-related toxic molecules or diabetes, one of the main target diseases for cell therapy due to their well-known healing impairments. METHODS: Cord blood-derived CD34+ cells were seeded in endothelial growth culture medium (EGM2) and ECFC colonies were obtained after 14-21 days. ECFC were exposed at pH 6.6 (preconditioned) or pH 7.4 (nonpreconditioned) for 6 h, and then pH was restored at 7.4. A model of type 2 diabetes induced by a high-fat and high-sucrose diet was developed in nude mice and hind limb ischemia was induced in these animals by femoral artery ligation. A P value < 0.05 was considered statistically significant (by one-way analysis of variance). RESULTS: We found that acidic preconditioning increased ECFC adhesion and the release of pro-angiogenic molecules, and protected ECFC from the cytotoxic effects of monosodium urate crystals, histones, and tumor necrosis factor (TNF)α, which induced necrosis, pyroptosis, and apoptosis, respectively. Noncytotoxic concentrations of high glucose, TNFα, or their combination reduced ECFC proliferation, stromal cell-derived factor (SDF)1-driven migration, and tubule formation on a basement membrane matrix, whereas almost no inhibition was observed in preconditioned ECFC. In type 2 diabetic mice, intravenous administration of preconditioned ECFC significantly induced blood flow recovery at the ischemic limb as measured by Doppler, compared with the phosphate-buffered saline (PBS) and nonpreconditioned ECFC groups. Moreover, the histologic analysis of gastrocnemius muscles showed an increased vascular density and reduced signs of inflammation in the animals receiving preconditioned ECFC. CONCLUSIONS: Acidic preconditioning improved ECFC survival and angiogenic activity in the presence of proinflammatory and damage signals present in the ischemic milieu, even under high glucose conditions, and increased their therapeutic potential for postischemia tissue regeneration in a murine model of type 2 diabetes. Collectively, our data suggest that acidic preconditioning of ECFC is a simple and inexpensive strategy to improve the effectiveness of cell transplantation in diabetes, where tissue repair is highly compromised.


Asunto(s)
Ácidos/química , Diabetes Mellitus Experimental/tratamiento farmacológico , Células Progenitoras Endoteliales/metabolismo , Glucosa/metabolismo , Neovascularización Fisiológica/fisiología , Animales , Diferenciación Celular , Proliferación Celular , Diabetes Mellitus Experimental/patología , Modelos Animales de Enfermedad , Humanos , Masculino , Ratones , Ratones Desnudos
9.
Stem Cell Rev Rep ; 14(1): 82-91, 2018 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-28980146

RESUMEN

Egfl7 (VE-statin) is a secreted protein mostly specific to the endothelial lineage during development and in the adult and which expression is enhanced during angiogenesis. Egfl7 involvement in human postnatal vasculogenesis remains unresolved yet. Our aim was to assess Egfl7 expression in several angiogenic cell types originating from human bone marrow, peripheral blood, or cord blood. We found that only endothelial colony forming cells (ECFC), which are currently considered as the genuine endothelial precursor cells, expressed large amounts of Egfl7. In order to assess its potential roles in ECFC, Egfl7 was repressed in ECFC by RNA interference and ECFC angiogenic capacities were tested in vitro and in vivo. Cell proliferation, differentiation, and migration were significantly improved when Egfl7 was repressed in ECFC in vitro, whereas miR-126-3p levels remained unchanged. In vivo, repression of Egfl7 in ECFC significantly improved post-ischemic revascularization in a model of mouse hind-limb ischemia. In conclusion, ECFC are the sole postnatal angiogenic cells which express large amounts of Egfl7 and whose angiogenic properties are repressed by this factor. Thus, Egfl7 inhibition may be considered as a therapeutic option to improve ECFC-mediated postnatal vasculogenesis and to optimize in vitro ECFC expansion in order to develop an optimized cell therapy approach.


Asunto(s)
Factores de Crecimiento Endotelial/metabolismo , Células Progenitoras Endoteliales/citología , Diferenciación Celular/fisiología , Movimiento Celular/genética , Movimiento Celular/fisiología , Células Cultivadas , Factores de Crecimiento Endotelial/genética , Células Progenitoras Endoteliales/metabolismo , Humanos , MicroARNs/genética , MicroARNs/metabolismo , Neovascularización Fisiológica/fisiología , Interferencia de ARN
10.
Thromb Haemost ; 117(10): 1908-1918, 2017 10 05.
Artículo en Inglés | MEDLINE | ID: mdl-28771278

RESUMEN

Endothelial colony-forming cells (ECFCs) are progenitor cells committed to endothelial lineages and have robust vasculogenic properties. Mesenchymal stem cells (MSCs) have been described to support ECFC-mediated angiogenic processes in various matrices. However, MSC-ECFC interactions in hind limb ischemia (HLI) are largely unknown. Here we examined whether co-administration of ECFCs and MSCs bolsters vasculogenic activity in nude mice with HLI. In addition, as we have previously shown that endoglin is a key adhesion molecule, we evaluated its involvement in ECFC/MSC interaction. Foot perfusion increased on day 7 after ECFC injection and was even better at 14 days. Co-administration of MSCs significantly increased vessel density and foot perfusion on day 7 but the differences were no longer significant at day 14. Analysis of mouse and human CD31, and in situ hybridization of the human ALU sequence, showed enhanced capillary density in ECFC+MSC mice. When ECFCs were silenced for endoglin, coinjection with MSCs led to lower vessel density and foot perfusion at both 7 and 14 days (p<0.001). Endoglin silencing in ECFCs did not affect MSC differentiation into perivascular cells or other mesenchymal lineages. Endoglin silencing markedly inhibited ECFC adhesion to MSCs. Thus, MSCs, when combined with ECFCs, accelerate muscle recovery in a mouse model of hind limb ischemia, through an endoglin-dependent mechanism.


Asunto(s)
Endoglina/metabolismo , Células Progenitoras Endoteliales/trasplante , Isquemia/cirugía , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas , Músculo Esquelético/irrigación sanguínea , Neovascularización Fisiológica , Adipogénesis , Animales , Adhesión Celular , Células Cultivadas , Condrogénesis , Modelos Animales de Enfermedad , Endoglina/genética , Células Progenitoras Endoteliales/metabolismo , Miembro Posterior , Isquemia/metabolismo , Isquemia/patología , Isquemia/fisiopatología , Masculino , Células Madre Mesenquimatosas/metabolismo , Ratones Desnudos , Músculo Esquelético/patología , Necrosis , Fenotipo , Interferencia de ARN , Recuperación de la Función , Flujo Sanguíneo Regional , Transducción de Señal , Factores de Tiempo , Transfección
11.
Cancer Lett ; 395: 11-19, 2017 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-28263839

RESUMEN

We previously reported that OPG is involved in ischemic tissue neovascularization through the secretion of SDF-1 by pretreated-OPG endothelial colony-forming cells (ECFCs). As the vascularization is one of the key factor influencing the tumour growth and cancer cell dissemination, we investigated whether OPG was able to modulate the invasion of human MNNG-HOS osteosarcoma and DU145 prostate cancer cell lines in vitro and in vivo. Cell motility was analysed in vitro by using Boyden chambers. Human GFP-labelled MMNG-HOS cells were inoculated in immunodeficient mice and the tumour nodules formed were then injected with OPG and/or FGF-2, AMD3100 or 0.9% NaCl (control group). Tumour growth was manually followed and angiogenesis was assessed by immunohistochemistry. In vitro, SDF-1 released by OPG-pretreated ECFCs markedly attracted both MNNG-HOS and DU145 cells and induced spontaneous migration of cancer cells. In vivo, tumour volumes were significantly increased in OPG-treated group compared to the control group and OPG potentiated the effect of FGF-2. Concomitantly, OPG alone or combined with FGF-2 increased the number of new vasculature compared to the control group. Interestingly AMD3100, an inhibitor of SDF-1, prevented the in vivo effects of OPG induced by SDF-1 This study provides experimental evidence that OPG promotes tumour development trough SDF-1/CXCR4 axis.


Asunto(s)
Movimiento Celular/efectos de los fármacos , Quimiocina CXCL12/fisiología , Neoplasias/etiología , Neovascularización Patológica/inducido químicamente , Osteoprotegerina/farmacología , Receptores CXCR4/fisiología , Animales , Línea Celular Tumoral , Factor 2 de Crecimiento de Fibroblastos/farmacología , Humanos , Ratones , Neoplasias/irrigación sanguínea , Neoplasias/patología , Ensayos Antitumor por Modelo de Xenoinjerto
12.
Circ Res ; 119(8): e110-26, 2016 Sep 30.
Artículo en Inglés | MEDLINE | ID: mdl-27582371

RESUMEN

RATIONALE: Sphingosine-1-phosphate (S1P) signaling is essential for vascular development and postnatal vascular homeostasis. The relative importance of S1P sources sustaining these processes remains unclear. OBJECTIVE: To address the level of redundancy in bioactive S1P provision to the developing and mature vasculature. METHODS AND RESULTS: S1P production was selectively impaired in mouse platelets, erythrocytes, endothelium, or smooth muscle cells by targeted deletion of genes encoding sphingosine kinases -1 and -2. S1P deficiency impaired aggregation and spreading of washed platelets and profoundly reduced their capacity to promote endothelial barrier function ex vivo. However, and in contrast to recent reports, neither platelets nor any other source of S1P was essential for vascular development, vascular integrity, or hemostasis/thrombosis. Yet rapid and profound depletion of plasma S1P during systemic anaphylaxis rendered both platelet- and erythrocyte-derived S1P essential for survival, with a contribution from blood endothelium observed only in the absence of circulating sources. Recovery was sensitive to aspirin in mice with but not without platelet S1P, suggesting that platelet activation and stimulus-response coupling is needed. S1P deficiency aggravated vasoplegia in this model, arguing a vital role for S1P in maintaining vascular resistance during recovery from circulatory shock. Accordingly, the S1P2 receptor mediated most of the survival benefit of S1P, whereas the endothelial S1P1 receptor was dispensable for survival despite its importance for maintaining vascular integrity. CONCLUSIONS: Although source redundancy normally secures essential S1P signaling in developing and mature blood vessels, profound depletion of plasma S1P renders both erythrocyte and platelet S1P pools necessary for recovery and high basal plasma S1P levels protective during anaphylactic shock.


Asunto(s)
Anafilaxia/metabolismo , Plaquetas/metabolismo , Endotelio Vascular/metabolismo , Eritrocitos/metabolismo , Homeostasis/fisiología , Lisofosfolípidos/deficiencia , Esfingosina/análogos & derivados , Anafilaxia/patología , Animales , Vasos Sanguíneos/crecimiento & desarrollo , Vasos Sanguíneos/metabolismo , Vasos Sanguíneos/patología , Endotelio Vascular/crecimiento & desarrollo , Endotelio Vascular/patología , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Humanos , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Transgénicos , Esfingosina/deficiencia
13.
Blood ; 126(15): 1823-30, 2015 Oct 08.
Artículo en Inglés | MEDLINE | ID: mdl-26316625

RESUMEN

Molecules that reduce the level of cyclic adenosine 5'-monophosphate (cAMP) in the platelet cytosol, such as adenosine 5'-diphosphate (ADP) secreted from dense granules, trigger platelet activation. Therefore, any change in the distribution and/or availability of cyclic nucleotides or ADP may interfere with platelet reactivity. In this study, we evaluated the role of multidrug resistance protein 4 (MRP4, or ABCC4), a nucleotide transporter, in platelet functions in vivo and in vitro by investigating MRP4-deficient mice. MRP4 deletion resulted in a slight increase in platelet count but had no impact on platelet ultrastructure. In MRP4-deficient mice, the arterial occlusion was delayed and the tail bleeding time was prolonged. In a model of platelet depletion and transfusion mimicking a platelet-specific knockout, mice injected with MRP4(-/-) platelets also showed a significant increase in blood loss compared with mice injected with wild-type platelets. Defective thrombus formation and platelet activation were confirmed in vitro by studying platelet adhesion to collagen in flow conditions, integrin αIIbß3 activation, washed platelet secretion, and aggregation induced by low concentrations of proteinase-activated receptor 4-activating peptide, U46619, or ADP. We found no role of MRP4 in ADP dense-granule storage, but MRP4 redistributed cAMP from the cytosol to dense granules, as confirmed by increased vasodilator-stimulated phosphoprotein phosphorylation in MRP4-deficient platelets. These data suggest that MRP4 promotes platelet aggregation by modulating the cAMP-protein kinase A signaling pathway, suggesting that MRP4 might serve as a target for novel antiplatelet agents.


Asunto(s)
Plaquetas/patología , AMP Cíclico/metabolismo , Homeostasis/fisiología , Proteínas Asociadas a Resistencia a Múltiples Medicamentos/fisiología , Activación Plaquetaria , Trombosis/patología , Animales , Transporte Biológico , Plaquetas/metabolismo , Células Cultivadas , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Ratones , Ratones Noqueados , Fosforilación , Agregación Plaquetaria , Pruebas de Función Plaquetaria , Transducción de Señal , Trombosis/metabolismo
14.
Int J Cancer ; 137(1): 73-85, 2015 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-25471534

RESUMEN

Interleukin-34 (IL-34) was recently characterized as the M-CSF "twin" cytokine, regulating the proliferation/differentiation/survival of myeloid cells. The implication of M-CSF in oncology was initially suspected by the reduced metastatic dissemination in knock-out mice, due to angiogenesis impairment. Based on this observation, our work studied the involvement of IL-34 in the pathogenesis of osteosarcoma. The in vivo effects of IL-34 were assessed on tissue vasculature and macrophage infiltration in a murine preclinical model based on a paratibial inoculation of human osteosarcoma cells overexpressing or not IL-34 or M-CSF. In vitro investigations using endothelial cell precursors and mature HUVEC cells were performed to analyse the involvement of IL-34 in angiogenesis and myeloid cell adhesion. The data revealed that IL-34 overexpression was associated with the progression of osteosarcoma (tumor growth, lung metastases) and an increase of neo-angiogenesis. In vitro analyses demonstrated that IL-34 stimulated endothelial cell proliferation and vascular cord formation. Pre-treatment of endothelial cells by chondroitinases/heparinases reduced the formation of vascular tubes and abolished the associated cell signalling. In addition, IL-34 increased the in vivo recruitment of M2 tumor-associated macrophages into the tumor tissue. IL-34 increased in vitro monocyte/CD34(+) cell adhesion to activated HUVEC monolayers under physiological shear stress conditions. This work also demonstrates that IL-34 is expressed by osteosarcoma cells, is regulated by TNF-α, IL-1ß, and contributes to osteosarcoma growth by increasing the neo-angiogenesis and the recruitment of M2 macrophages. By promoting new vessel formation and extravasation of immune cells, IL-34 may play a key role in tumor development and inflammatory diseases.


Asunto(s)
Neoplasias Óseas/patología , Interleucinas/metabolismo , Neoplasias Pulmonares/patología , Macrófagos/metabolismo , Neovascularización Patológica/metabolismo , Osteosarcoma/patología , Animales , Neoplasias Óseas/irrigación sanguínea , Neoplasias Óseas/metabolismo , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Regulación Neoplásica de la Expresión Génica , Células Endoteliales de la Vena Umbilical Humana , Humanos , Técnicas In Vitro , Interleucina-1beta/metabolismo , Interleucinas/farmacología , Neoplasias Pulmonares/irrigación sanguínea , Neoplasias Pulmonares/metabolismo , Ratones , Trasplante de Neoplasias , Osteosarcoma/irrigación sanguínea , Osteosarcoma/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo
15.
Angiogenesis ; 17(4): 867-79, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-24854678

RESUMEN

OBJECTIVE: Acidosis is present in several pathological conditions where vasculogenesis takes place including ischemia, tumor growth and wound healing. We have previously demonstrated that acidosis induces human CD34+ cell apoptosis. Considering that endothelial colony-forming cells (ECFC) are a subpopulation of CD34+ cells and key players in vasculogenesis, in the present study we investigated the effect of acidosis on the survival and functionality of ECFC. APPROACH AND RESULTS: Endothelial colony-forming cells obtained by differentiation of human cord blood CD34+ cells in endothelial growth medium-2 for 14-21 days were exposed at pH 7.4, 7.0 or 6.6. We found that acidosis failed to induce ECFC apoptosis and, although an early reduction in proliferation, chemotaxis, wound healing and capillary-like tubule formation was observed, once the medium pH was restored to 7.4, ECFC proliferation and tubulogenesis were augmented. Stromal cell derived factor-1 (SDF1)-driven migration and chemokine receptor type 4 surface expression were also increased. The maximal proangiogenic effect exerted by acidic preconditioning was observed after 6 h at pH 6.6. Furthermore, preconditioned ECFC showed an increased ability to promote tissue revascularization in a murine model of hind limb ischemia. Immunoblotting assays showed that acidosis activated AKT and ERK1/2 and inhibited p38 pathways. Proliferation rises triggered by acidic preconditioning were no longer observed after AKT or ERK1/2 inhibition, whereas p38 suppression not only mimicked but also potentiated the effect of acidosis on ECFC tubule formation abilities. CONCLUSIONS: These results demonstrate that acidic preconditioning greatly increases ECFC-mediated angiogenesis in vitro including ECFC proliferation, tubulogenesis and SDF1-driven chemotaxis and is a positive regulator of microvessel formation in vivo.


Asunto(s)
Ácidos/química , Medios de Cultivo/química , Células Endoteliales/citología , Células Madre/citología , Animales , Antígenos CD34/metabolismo , Apoptosis , Ciclo Celular , Diferenciación Celular , Proliferación Celular , Quimiotaxis , Humanos , Concentración de Iones de Hidrógeno , Isquemia/patología , Masculino , Ratones , Ratones Desnudos , Neovascularización Fisiológica/efectos de los fármacos , Cicatrización de Heridas
16.
Angiogenesis ; 17(3): 603-16, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24419917

RESUMEN

OBJECTIVES: We studied whether plasma levels of angiogenic factors VEGF and placental growth factor (PlGF) in coronary artery disease patients or undergoing cardiac surgery are modified, and whether those factors modulate endothelial progenitor's angiogenic potential. METHODS AND RESULTS: A total of 143 patients' plasmas from two different studies were analyzed (30 coronary artery disease patients, 30 patients with stable angina, coupled with 30 age and sex-matched controls; 53 patients underwent cardiac surgery). Among factors screened, only PlGF was found significantly increased in these pathological populations. PlGF-1 and PlGF-2 were then tested on human endothelial-colony-forming cells (ECFCs). We found that PlGF-1 and PlGF-2 induce VEGFR1 phosphorylation and potentiate ECFCs tubulogenesis in vitro. ECFCs VEGFR1 was further inhibited using a specific small interfering RNA (siRNA) and the chemical compound 4321. We then observed that the VEGFR1-siRNA and the compound 4321 decrease ECFCs tubulogenesis potential in vitro. Finally, we tested the compound 4321 in the preclinical Matrigel(®)-plug model with C57Bl/6J mice as well as in the murine hindlimb ischemia model. We found that 4321 inhibited the plug vascularization, attested by the hemoglobin content and the VE-Cadherin expression level and that 4321 inhibited the post-ischemic revascularization. CONCLUSION: PlGF plasma levels were found increased in cardiovascular patients. Disrupting PlGF/VEGFR1 pathway could modulate ECFC-induced tubulogenesis, the cell type responsible for newly formed vessels in vivo.


Asunto(s)
Diferenciación Celular , Células Endoteliales/metabolismo , Células Madre/metabolismo , Receptor 1 de Factores de Crecimiento Endotelial Vascular/metabolismo , Animales , Procedimientos Quirúrgicos Cardíacos , Diferenciación Celular/efectos de los fármacos , Ensayos de Migración Celular , Proliferación Celular/efectos de los fármacos , Colágeno/metabolismo , Ensayo de Unidades Formadoras de Colonias , Enfermedad de la Arteria Coronaria/sangre , Enfermedad de la Arteria Coronaria/patología , Combinación de Medicamentos , Células Endoteliales/efectos de los fármacos , Miembro Posterior/irrigación sanguínea , Miembro Posterior/patología , Humanos , Isquemia/patología , Laminina/metabolismo , Proteínas de la Membrana/sangre , Ratones Endogámicos C57BL , Neovascularización Fisiológica/efectos de los fármacos , Fosforilación/efectos de los fármacos , Proteoglicanos/metabolismo , ARN Interferente Pequeño/metabolismo , Proteínas Recombinantes/farmacología , Células Madre/efectos de los fármacos , Factor A de Crecimiento Endotelial Vascular/sangre
17.
Cytotherapy ; 16(2): 213-24, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24280239

RESUMEN

BACKGROUND AIMS: Circulating endothelial progenitor cells and especially endothelial colony-forming cells (ECFCs) are promising candidate cells for endothelial regenerative medicine of ischemic diseases, but the conditions for an optimal collection from adult blood must be improved. METHODS: On the basis of a recently reported vascular niche of ECFCs, we hypothesized that a local ischemia could trigger ECFC mobilization from the vascular wall into peripheral blood to optimize their collection for autologous implantation in critical leg ischemia. Because the target population with critical leg ischemia is composed of elderly patients in whom a vascular impairment has been documented, we also analyzed the impact of aging on ECFC mobilization and vascular integrity. RESULTS: After having defined optimized ECFC culture conditions, we studied the effect of forearm ischemia on ECFC numbers and functions in 26 healthy volunteers (13 volunteers ages 20-30-years old versus 13 volunteers ages 60-70 years old). The results show that forearm ischemia induced an efficient local ischemia and a normal endothelial response but did not mobilize ECFCs regardless of the age group. Moreover, we report an alteration of angiogenic properties of ECFCs obtained after forearm ischemia, in vitro as well as in vivo in a hindlimb ischemia murine model. This impaired ECFC angiogenic potential was not associated with a quantitative modification of the circulating endothelial compartment. CONCLUSIONS: The procedure of local ischemia, although reulting in a preserved endothelial reactivity, did not mobilize ECFCs but altered their angiogenic potential.


Asunto(s)
Células Madre Adultas/metabolismo , Células Endoteliales/metabolismo , Antebrazo/fisiopatología , Miembro Posterior/fisiopatología , Isquemia/fisiopatología , Adulto , Células Madre Adultas/patología , Anciano , Animales , Diferenciación Celular , Células Cultivadas , Células Endoteliales/patología , Células Endoteliales/trasplante , Femenino , Antebrazo/irrigación sanguínea , Miembro Posterior/irrigación sanguínea , Humanos , Isquemia/patología , Masculino , Ratones , Ratones Desnudos , Persona de Mediana Edad , Neovascularización Fisiológica , Trasplante de Células Madre , Células Madre , Adulto Joven
18.
Tissue Eng Part A ; 20(1-2): 275-84, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24059447

RESUMEN

Human adipose-derived stromal cells (hASCs) may hold potential for bone tissue engineering. Osteogenic differentiation of these cells is crucial to bone formation. Low-molecular-weight fucoidan (LMWF) is a sulfated polysaccharide that potentiates several growth factors, including pro-angiogenic growth factors. To investigate whether hASC preconditioning with LMWF promoted bone repair, we compared the effects of LMWF and low-molecular-weight heparin on hASC phenotype and osteogenic differentiation. LMWF did not modify the stem-cell phenotype of hASCs but enhanced their osteogenic differentiation (formation of calcium deposits, increased activity and expression of alkaline phosphatase, and increased expression of osteopontin and runt-related transcription factor 2). However, when hASCs were exposed to LMWF before their adhesion to biphasic calcium phosphate particles and implantation in a bone-growth mouse model, no bone formation was apparent after 5 or 8 weeks, probably due to cell death. In conclusion, LMWF may hold promise for enhancing the osteogenic differentiation of hASCs before their implantation. However, concomitant vascularization would be required to enhance bone formation.


Asunto(s)
Tejido Adiposo/citología , Osteogénesis/efectos de los fármacos , Polisacáridos/química , Polisacáridos/farmacología , Animales , Diferenciación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Humanos , Hidroxiapatitas/farmacología , Masculino , Ratones , Modelos Animales , Peso Molecular , Fenotipo , Células del Estroma/citología , Células del Estroma/efectos de los fármacos , Células del Estroma/metabolismo
19.
Cardiovasc Res ; 95(1): 39-47, 2012 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-22517984

RESUMEN

AIMS: Integrins α6ß1 and α6ß4 are receptors for laminins, the main components of the basement membrane underlying the endothelial cells. In vitro, α6 integrin subunit (α6) expression at the surface of endothelial cells and their progenitors (EPCs) is up-regulated by pro-angiogenic growth factors and is crucial for adhesion, migration, and pseudotube formation. We investigated the role for α6 in post-ischaemic vascular repair in vivo. METHODS AND RESULTS: We used the cre-lox system to generate a mouse line with specific α6 gene deletion in Tie2-lineage cells. In a model of hind-limb ischaemia, Tie2-dependent α6 deletion reduced neovessel formation and reperfusion of the ischaemic limb. Concerning the role for α6 in post-ischaemic vasculogenesis, we showed previously that α6 was required for EPC recruitment at the site of ischaemia. Here, we found that α6 deletion also reduced EPC mobilization from the bone marrow after ischaemia. Examination of the ischaemic muscles showed that Tie2-dependent α6 deletion decreased the recruitment of pro-angiogenic Tie2-expressing macrophages. In the Matrigel plug assay, fibroblast growth factor-2-induced vascularization was diminished in mice lacking endothelial α6. To specifically investigate the role for α6 in angiogenesis, aortic rings were embedded in Matrigel or collagen and cultured ex vivo. In Matrigel, neovessel outgrowth from rings lacking α6 was strongly diminished, whereas no genotype-dependent difference occurred for rings in collagen. CONCLUSION: α6 plays a major role in both post-ischaemic angiogenesis and vasculogenesis.


Asunto(s)
Integrina alfa6/fisiología , Isquemia/fisiopatología , Neovascularización Fisiológica , Proteínas Tirosina Quinasas Receptoras/fisiología , Animales , Linaje de la Célula , Movimiento Celular , Factor 2 de Crecimiento de Fibroblastos/farmacología , Masculino , Ratones , Ratones Noqueados , Subunidades de Proteína/fisiología , Receptor TIE-2
20.
Blood ; 118(6): 1699-709, 2011 Aug 11.
Artículo en Inglés | MEDLINE | ID: mdl-21659549

RESUMEN

Low birth weight (LBW) is associated with increased risk of cardiovascular diseases at adulthood. Nevertheless, the impact of LBW on the endothelium is not clearly established. We investigate whether LBW alters the angiogenic properties of cord blood endothelial colony forming cells (LBW-ECFCs) in 25 preterm neonates compared with 25 term neonates (CT-ECFCs). We observed that LBW decreased the number of colonies formed by ECFCs and delayed the time of appearance of their clonal progeny. LBW dramatically reduced LBW-ECFC capacity to form sprouts and tubes, to migrate and to proliferate in vitro. The angiogenic defect of LBW-ECFCs was confirmed in vivo by their inability to form robust capillary networks in Matrigel plugs injected in nu/nu mice. Gene profile analysis of LBW-ECFCs demonstrated an increased expression of antiangiogenic genes. Among them, thrombospondin 1 (THBS1) was highly expressed at RNA and protein levels in LBW-ECFCs. Silencing THBS1 restored the angiogenic properties of LBW-ECFCs by increasing AKT phosphorylation. The imbalance toward an angiostatic state provide a mechanistic link between LBW and the impaired angiogenic properties of ECFCs and allows the identification of THBS1 as a novel player in LBW-ECFC defect, opening new perspectives for novel deprogramming agents.


Asunto(s)
Células Endoteliales/metabolismo , Perfilación de la Expresión Génica , Recién Nacido de Bajo Peso/sangre , Recien Nacido Prematuro/sangre , Neovascularización Fisiológica/genética , Células Madre/metabolismo , Animales , Vasos Sanguíneos/citología , Vasos Sanguíneos/crecimiento & desarrollo , Vasos Sanguíneos/metabolismo , Western Blotting , Proliferación Celular , Células Cultivadas , Células Endoteliales/citología , Femenino , Sangre Fetal/citología , Humanos , Recién Nacido , Masculino , Ratones , Ratones Endogámicos , Ratones Desnudos , Fosforilación , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Interferencia de ARN , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Trasplante de Células Madre/métodos , Células Madre/citología , Trombospondina 1/genética , Trombospondina 1/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...