Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Cell Mol Life Sci ; 79(3): 145, 2022 Feb 21.
Artículo en Inglés | MEDLINE | ID: mdl-35190870

RESUMEN

Hemophilia A is an inherited X-linked recessive bleeding disorder caused by deficient activity of blood coagulation factor VIII (FVIII). In addition, hemophilia patients show associated diseases including osteopenia, altered inflammation and vascular fragility which may represent the consequence of recurrent bleeding or may be related to the direct FVIII deficiency. Nowadays, recombinant FVIII is proposed to treat hemophilia patients with no circulating FVIII inhibitor. Initially described as a coenzyme to factor IXa for initiating thrombin generation, there is emerging evidence that FVIII is involved in multiple biological systems, including bone, vascular and immune systems. The present study investigated: (i) the functional activities of recombinant human FVIII (rFVIII) on endothelial cells, and (ii) the impact of rFVIII activities on the functional interactions of human monocytes and endothelial cells. We then investigated whether rFVIII had a direct effect on the adhesion of monocytes to the endothelium under physiological flow conditions. We observed that direct biological activities for rFVIII in endothelial cells were characterized by: (i) a decrease in endothelial cell adhesion to the underlying extracellular matrix; (ii) regulation of the transcriptomic and protein profiles of endothelial cells; (iii) an increase in the vascular tubes formed and vascular permeability in vitro; and (iv) an increase in monocyte adhesion activated endothelium and transendothelial migration. By regulating vascular permeability plus leukocyte adhesion and transendothelial migration, the present work highlights new biological functions for FVIII.


Asunto(s)
Permeabilidad de la Membrana Celular , Endotelio Vascular/metabolismo , Factor VIII/metabolismo , Macrófagos/metabolismo , Neovascularización Fisiológica , Adhesión Celular , Movimiento Celular , Endotelio Vascular/citología , Factor VIII/genética , Humanos , Macrófagos/citología , Proteoma , Transcriptoma
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.
J Transl Med ; 17(1): 261, 2019 08 09.
Artículo en Inglés | MEDLINE | ID: mdl-31399109

RESUMEN

BACKGROUND: Cell therapy has been proposed for patients with critical limb ischemia (CLI). Autologous bone marrow derived cells (BMCs) have been mostly used, mesenchymal stem cells (MSCs) being an alternative. The aim of this study was to characterize two types of MSCs and evaluate their efficacy. METHODS: MSCs were obtained from CLI-patients BMCs. Stimulated- (S-) MSCs were cultured in endothelial growth medium. Cells were characterized by the expression of cell surface markers, the relative expression of 6 genes, the secretion of 10 cytokines and the ability to form vessel-like structures. The cell proangiogenic properties was analysed in vivo, in a hindlimb ischemia model. Perfusion of lower limbs and functional tests were assessed for 28 days after cell infusion. Muscle histological analysis (neoangiogenesis, arteriogenesis and muscle repair) was performed. RESULTS: S-MSCs can be obtained from CLI-patients BMCs. They do not express endothelial specific markers but can be distinguished from MSCs by their secretome. S-MSCs have the ability to form tube-like structures and, in vivo, to induce blood flow recovery. No amputation was observed in S-MSCs treated mice. Functional tests showed improvement in treated groups with a superiority of MSCs and S-MSCs. In muscles, CD31+ and αSMA+ labelling were the highest in S-MSCs treated mice. S-MSCs induced the highest muscle repair. CONCLUSIONS: S-MSCs exert angiogenic potential probably mediated by a paracrine mechanism. Their administration is associated with flow recovery, limb salvage and muscle repair. The secretome from S-MSCs or secretome-derived products may have a strong potential in vessel regeneration and muscle repair. Trial registration NCT00533104.


Asunto(s)
Medios de Cultivo/farmacología , Células Endoteliales/citología , Extremidades/irrigación sanguínea , Isquemia/terapia , Células Madre Mesenquimatosas/citología , Adulto , Anciano , Animales , Arterias/crecimiento & desarrollo , Células Cultivadas , Células Endoteliales/efectos de los fármacos , Extremidades/patología , Femenino , Miembro Posterior/irrigación sanguínea , Humanos , Isquemia/patología , Masculino , Trasplante de Células Madre Mesenquimatosas , Ratones Endogámicos BALB C , Ratones Desnudos , Persona de Mediana Edad , Músculos/irrigación sanguínea , Músculos/patología , Neovascularización Fisiológica , Organogénesis , Flujo Sanguíneo Regional
5.
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
6.
Small ; 14(16): e1800020, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29542273

RESUMEN

Once injected into a living organism, cells diffuse or migrate around the initial injection point and become impossible to be visualized and tracked in vivo. The present work concerns the development of a new technique for therapeutic cell labeling and subsequent in vivo visualization and magnetic retention. It is hypothesized and subsequently demonstrated that nanohybrids made of persistent luminescence nanoparticles and ultrasmall superparamagnetic iron oxide nanoparticles incorporated into a silica matrix can be used as an effective nanoplatform to label therapeutic cells in a nontoxic way in order to dynamically track them in real-time in vitro and in living mice. As a proof-of-concept, it is shown that once injected, these labeled cells can be visualized and attracted in vivo using a magnet. This first step suggests that these nanohybrids represent efficient multifunctional nanoprobes for further imaging guided cell therapies development.


Asunto(s)
Nanopartículas/química , Compuestos Férricos/química , Luminiscencia
7.
J Cell Mol Med ; 21(2): 349-363, 2017 02.
Artículo en Inglés | MEDLINE | ID: mdl-27641937

RESUMEN

This study aimed at characterizing the impact of type 2 diabetes mellitus (T2DM) on the bone marrow mesenchymal stem cell (BMMSC) secretome and angiogenic properties. BMMSCs from Zucker diabetic fatty rats (ZDF) (a T2DM model) and Zucker LEAN littermates (control) were cultured. The supernatant conditioned media (CM) from BMMSCs of diabetic and control rats were collected and analysed. Compared to results obtained using CM from LEAN-BMMSCs, the bioactive content of ZDF-BMMSC CM (i) differently affects endothelial cell (HUVEC) functions in vitro by inducing increased (3.5-fold; P < 0.01) formation of tubule-like structures and migration of these cells (3-fold; P < 0.001), as well as promotes improved vascular formation in vivo, and (ii) contains different levels of angiogenic factors (e.g. IGF1) and mediators, such as OSTP, CATD, FMOD LTBP1 and LTBP2, which are involved in angiogenesis and/or extracellular matrix composition. Addition of neutralizing antibodies against IGF-1, LTBP1 or LTBP2 in the CM of BMMSCs from diabetic rats decreased its stimulatory effect on HUVEC migration by approximately 60%, 40% or 40%, respectively. These results demonstrate that BMMSCs from T2DM rats have a unique secretome with distinct angiogenic properties and provide new insights into the role of BMMSCs in aberrant angiogenesis in the diabetic milieu.


Asunto(s)
Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Células Madre Mesenquimatosas/metabolismo , Neovascularización Fisiológica , Proteoma/metabolismo , Animales , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Medios de Cultivo Condicionados/farmacología , Diabetes Mellitus Experimental/genética , Diabetes Mellitus Experimental/patología , Diabetes Mellitus Tipo 2/genética , Diabetes Mellitus Tipo 2/patología , Proteínas de la Matriz Extracelular/metabolismo , Perfilación de la Expresión Génica , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Factor I del Crecimiento Similar a la Insulina/farmacología , Masculino , Células Madre Mesenquimatosas/efectos de los fármacos , Ratones Desnudos , Neovascularización Fisiológica/efectos de los fármacos , Neovascularización Fisiológica/genética , Proteómica , Ratas Zucker , Especies Reactivas de Oxígeno/metabolismo
8.
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
9.
Mar Drugs ; 13(7): 4156-70, 2015 Jun 30.
Artículo en Inglés | MEDLINE | ID: mdl-26133555

RESUMEN

Critical limb ischemia (CLI) induces the secretion of paracrine signals, leading to monocyte recruitment and thereby contributing to the initiation of angiogenesis and tissue healing. We have previously demonstrated that fucoidan, an antithrombotic polysaccharide, promotes the formation of new blood vessels in a mouse model of hindlimb ischemia. We examined the effect of fucoidan on the capacity of peripheral blood monocytes to adhere and migrate. Monocytes negatively isolated with magnetic beads from peripheral blood of healthy donors were treated with fucoidan. Fucoidan induced a 1.5-fold increase in monocyte adhesion to gelatin (p < 0.05) and a five-fold increase in chemotaxis in Boyden chambers (p < 0.05). Fucoidan also enhanced migration 2.5-fold in a transmigration assay (p < 0.05). MMP9 activity in monocyte supernatants was significantly enhanced by fucoidan (p < 0.05). Finally, Western blot analysis of fucoidan-treated monocytes showed upregulation of ERK/p38 phosphorylation. Inhibition of ERK/p38 phosphorylation abrogated fucoidan enhancement of migration (p < 0.01). Fucoidan displays striking biological effects, notably promoting monocyte adhesion and migration. These effects involve the ERK and p38 pathways, and increased MMP9 activity. Fucoidan could improve critical limb ischemia by promoting monocyte recruitment.


Asunto(s)
Movimiento Celular/efectos de los fármacos , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Metaloproteinasa 9 de la Matriz/metabolismo , Monocitos/efectos de los fármacos , Polisacáridos/farmacología , Western Blotting , Adhesión Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Citometría de Flujo , Humanos , Integrinas/fisiología , Sistema de Señalización de MAP Quinasas/fisiología , Metaloproteinasa 9 de la Matriz/fisiología , Monocitos/fisiología , Receptores CCR2/fisiología
10.
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
11.
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
12.
Int J Biol Macromol ; 264(Pt 1): 130424, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38428772

RESUMEN

The fishing and aquaculture industries generate a huge amount of waste during processing and preservation operations, especially those of tuna. Recovering these by-products is a major economic and environmental challenge for manufacturers seeking to produce new active biomolecules of interest. A new hyaluronic acid was extracted from bluefin tuna's vitreous humour to assess its antioxidant and pharmacological activities. The characterization by infrared spectroscopy (FT-IR), nuclear magnetic resonance ((1D1H) and 2D (1H COSY, 1H/13C HSQC)) and size exclusion chromatography (SEC/MALS/DRI/VD) revealed that the extracted polysaccharide was a hyaluronic acid with high uronic acid content (55.8 %) and a weight average molecular weight of 888 kDa. This polymer possesses significant anti-radical activity and ferrous chelating capacity. In addition, pharmacological evaluation of its anti-inflammatory and analgesic potential, using preclinical models, in comparison with reference drugs (Dexamethasone, diclofenac, and acetylsalicylate of lysine), revealed promising anti-inflammatory activity as well as interesting peripheral and central antinociceptive activity. Therefore, our new hyaluronic acid compound may therefore serve as a potential drug candidate for the treatment of pain sensation and inflammation of various pathological origins.


Asunto(s)
Ácido Hialurónico , Atún , Animales , Espectroscopía Infrarroja por Transformada de Fourier , Polisacáridos/química , Antiinflamatorios/farmacología , Antiinflamatorios/química
13.
Angiogenesis ; 16(3): 575-93, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23386104

RESUMEN

Osteoprotegerin (OPG), a soluble tumour necrosis factor receptor superfamily member, inhibits RANKL-mediated osteoclastogenesis. We have previously reported that OPG enhances the proangiogenic properties of endothelial colony-forming cells (ECFCs) in vitro, and promotes vasculogenesis in vivo. Here we investigated how OPG promotes neovascularisation. Proteomic experiments showed that OPG pretreatment affected ECFCs protein expression in two ways, 23 spots being down-regulated and 6 upregulated. These spots corresponded to proteins involved in cell motility, adhesion, signal transduction and apoptosis. In keeping with these proteomic results, we found that OPG induced ECFCs adhesion to activated endothelium in shear stress conditions, promoting intermediate but not focal adhesion to fibronectin and collagen. Treatment with OPG induced a reorganization of the ECFCs cytoskeleton, with the emergence of cell protrusions characteristic of a migratory phenotype. These effects correlated with decreased FAK phosphorylation and enhanced integrin αVß3 expression. OPG drastically reduced caspase-3/7 activities and maintained ECFCs viability after 48 h of treatment. All these effects were significantly attenuated by ECFCs incubation with the CXCR4 antagonist AMD-3100, and by prior heparan sulphate proteoglycan disruption. The proangiogenic properties of OPG appeared to be mediated by the proteoglycan syndecan-1, although OPG 1-194 lacking its heparin-binding domain still had pro-vasculogenic effects in vitro and in vivo. These results suggest that OPG may interact with ECFCs by binding to HSPGs/syndecan-1, thereby induce an anti-adhesive effect and promoting ECFCs migration through a SDF-1/CXCR4 dependent pathway.


Asunto(s)
Células Endoteliales/efectos de los fármacos , Regulación de la Expresión Génica/efectos de los fármacos , Neovascularización Fisiológica/efectos de los fármacos , Osteoprotegerina/farmacología , Bencilaminas , Caspasa 3/metabolismo , Adhesión Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Ciclamas , Citoesqueleto/efectos de los fármacos , Células Endoteliales/fisiología , Quinasa 1 de Adhesión Focal/metabolismo , Regulación de la Expresión Génica/fisiología , Compuestos Heterocíclicos , Células Endoteliales de la Vena Umbilical Humana , Humanos , Integrina alfaVbeta3/metabolismo , Neovascularización Fisiológica/fisiología , Osteoprotegerina/metabolismo , Fosforilación/efectos de los fármacos , Proteómica , Sindecano-1/metabolismo
14.
Arterioscler Thromb Vasc Biol ; 31(3): 551-9, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21148423

RESUMEN

OBJECTIVE: We examined whether plasma levels of angiogenic factors are altered in plasma of patients with peripheral arterial disease (PAD) and whether these factors affect endothelial progenitor cell-induced angiogenesis. METHODS AND RESULTS: Plasma was collected from 184 patients with PAD and 330 age-matched healthy controls. Vascular endothelial growth factor and placental growth factor concentrations did not differ between the groups, whereas we found a linear correlation between PAD disease and thrombospondin (TSP)-1 plasma level. TSP-1 was expressed in newly formed vessels in PAD patients having received local injections of bone marrow mononuclear cells. To analyze the functional role of TSP-1 during neoangiogenesis, we used a Matrigel-plug assay and showed that vascularization of implanted Matrigel-plugs was increased in TSP-1(-/-) mice. Moreover, injections of TSP-1 in C57Bl6/J mice after hindlimb ischemia induced a significant decrease of blood flow recovery. To investigate the effects of TSP-1 on human endothelial colony-forming cell (ECFC) angiogenic potential, recombinant human TSP-1 and a small interfering RNA were used. In vitro, TSP-1 N-terminal part significantly enhanced ECFC adhesion, whereas recombinant human TSP-1 had a negative effect on ECFC angiogenic potential. This effect, mediated by CD47 binding, modulated stromal cell-derived factor 1/CXC chemokine receptor 4 pathway. CONCLUSIONS: TSP-1 is a potential biomarker of PAD and ECFC-induced angiogenesis, suggesting that TSP-1 modulation might improve local tissue ischemia in this setting. ( CLINICAL TRIAL REGISTRATION: NCT00377897.).


Asunto(s)
Proteínas Angiogénicas/sangre , Células Endoteliales/metabolismo , Isquemia/metabolismo , Músculo Esquelético/irrigación sanguínea , Neovascularización Fisiológica , Enfermedad Arterial Periférica/sangre , Células Madre/metabolismo , Trombospondina 1/sangre , Proteínas Angiogénicas/administración & dosificación , Proteínas Angiogénicas/deficiencia , Proteínas Angiogénicas/genética , Animales , Biomarcadores/sangre , Antígeno CD47/metabolismo , Estudios de Casos y Controles , Adhesión Celular , Proliferación Celular , Células Cultivadas , Quimiocina CXCL12/metabolismo , Modelos Animales de Enfermedad , Células Endoteliales/trasplante , Miembro Posterior , Humanos , Isquemia/fisiopatología , Isquemia/cirugía , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Enfermedad Arterial Periférica/fisiopatología , Enfermedad Arterial Periférica/cirugía , Fenotipo , Factor de Crecimiento Placentario , Proteínas Gestacionales/sangre , Interferencia de ARN , Receptores CXCR4/metabolismo , Trasplante de Células Madre , Trombospondina 1/administración & dosificación , Trombospondina 1/deficiencia , Trombospondina 1/genética , Resultado del Tratamiento , Factor A de Crecimiento Endotelial Vascular/sangre
15.
Arterioscler Thromb Vasc Biol ; 30(8): 1569-75, 2010 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-20508204

RESUMEN

OBJECTIVE: Alpha6 integrin subunit (alpha6) expression is increased by proangiogenic growth factors such as vascular endothelial growth factor (VEGF) and fibroblast growth factor. This increase correlates with enhanced in vitro tube formation by endothelial cells and their progenitors called endothelial colony-forming cells (ECFCs). We thus studied the role of alpha6 in vasculogenesis induced by human ECFCs, in a mouse model of hindlimb ischemia. METHODS AND RESULTS: We used small interfering RNA (siRNA) to inhibit alpha6 expression on the surface of ECFCs. For in vivo studies, human ECFCs were injected intravenously into a nude mouse model of unilateral hind limb ischemia. Transfection with siRNA alpha6 abrogated neovessel formation and reperfusion of the ischemic hind limb induced by ECFCs (P<0.01 and P<0.001, respectively). It also inhibited ECFC incorporation into the vasculature of the ischemic muscle (P<0.001). In vitro, siRNA alpha6 inhibited ECFC adhesion (P<0.01), pseudotube formation on Matrigel, migration, and AKT phosphorylation (P<0.0001), with no effect on cell proliferation or apoptosis. CONCLUSIONS: alpha6 Expression is required for ECFC migration, adhesion, recruitment at the site of ischemia, and the promotion of the postischemic vascular repair. Thus, we have demonstrated a major role of alpha6 in the proangiogenic properties of ECFCs.


Asunto(s)
Células Endoteliales/metabolismo , Integrina alfa6/metabolismo , Isquemia/metabolismo , Músculo Esquelético/irrigación sanguínea , Músculo Esquelético/metabolismo , Neovascularización Fisiológica , Células Madre/metabolismo , Animales , Apoptosis , Capilares/metabolismo , Capilares/fisiopatología , Adhesión Celular , Movimiento Celular , Proliferación Celular , Células Cultivadas , Modelos Animales de Enfermedad , Células Endoteliales/trasplante , Humanos , Integrina alfa6/genética , Isquemia/genética , Isquemia/patología , Isquemia/fisiopatología , Flujometría por Láser-Doppler , Masculino , Ratones , Ratones Desnudos , Músculo Esquelético/patología , Neovascularización Fisiológica/genética , Fosforilación , Proteínas Proto-Oncogénicas c-akt/metabolismo , Interferencia de ARN , Flujo Sanguíneo Regional , Trasplante de Células Madre , Transfección
16.
Arterioscler Thromb Vasc Biol ; 28(4): 644-50, 2008 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-18239152

RESUMEN

OBJECTIVE: As SDF-1 and its cognate receptor CXCR4 play a key role in the survival and mobilization of immature cells, we examined whether preconditioning of endothelial progenitor cells (EPCs) with SDF-1 could further promote their capacity to enhance angiogenesis. METHODS AND RESULTS: EPC exposure to 100 ng/mL SDF-1 for 30 min induced a proangiogenic phenotype, with cell migration and differentiation into vascular cords in Matrigel and increased their therapeutic potential in a nude mouse model of hindlimb ischemia. This pretreatment enhanced EPC adhesion to activated endothelium in physiological conditions of blood flow by stimulating integrin-mediated EPCs binding to endothelial cells. Pretreated EPCs showed significantly upregulated surface alpha4 and alphaM integrin subunit expression involved in the homing of immature cells to a neovasculature and enhanced FGF-2 and promatrix metalloproteinase (MMP)-2 secretion. All these effects were significantly attenuated by EPC incubation with AMD-3100, a CXCR4 antagonist, by prior HSPGs disruption and by HUVEC incubation with anti-intercellular adhesion molecule1 (ICAM-1) and anti-vascular cell adhesion molecule (VCAM) blocking antibodies. Pretreated EPCs adhered very rapidly (within minutes) and were resistant to shear stresses of up to 2500 s(-1). CONCLUSIONS: SDF-1 pretreatment during EPC expansion stimulates EPC adhesion to endothelial cells and thus augments the efficiency of cell therapy for ischemic vascular diseases.


Asunto(s)
Quimiocina CXCL12/farmacología , Trasplante de Células Madre de Sangre del Cordón Umbilical/métodos , Células Endoteliales/efectos de los fármacos , Células Endoteliales/trasplante , Neovascularización Fisiológica/efectos de los fármacos , Células Madre/efectos de los fármacos , Células Madre/fisiología , Animales , Quimiocina CXCL12/genética , Quimiocina CXCL12/metabolismo , Células Endoteliales/fisiología , Miembro Posterior , Humanos , Técnicas In Vitro , Isquemia/terapia , Ratones , Ratones Desnudos , ARN Mensajero/genética , ARN Mensajero/metabolismo , Receptores CXCR4/genética , Receptores CXCR4/metabolismo , Trasplante Heterólogo
17.
J Innate Immun ; 11(4): 330-346, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30557873

RESUMEN

Exposition of neutrophils (polymorphonuclear neutrophils, PMNs) to bacterial products triggers exacerbated activation of these cells, increasing their harmful effects on host tissues. We evaluated the possibility of interfering with the classic immune innate responses of human PMNs exposed to bacterial endotoxin (lipopolysaccharide, LPS), and further stimulated with bacterial formyl peptide (N-formyl-methionine-leucine-phenylalanine, fMLP). We showed that the low- molecular-weight fucoidan (LMW-Fuc), a polysaccharide extracted from brown algae, attenuated the exacerbated activation induced by fMLP on LPS-primed PMNs, in vitro, impairing chemotaxis, NET formation, and the pro-survival and pro-oxidative effects. LMW-Fuc also inhibited the activation of canonical signaling pathways, AKT, bad, p47phox and MLC, activated by the exposition of PMN to bacterial products. The activation of PMN by sequential exposure to LPS and fMLP induced the release of L-selectin+ microparticles, which were able to trigger extracellular reactive oxygen species production by fresh PMNs and macrophages. Furthermore, we observed that LMW-Fuc inhibited microparticle release from activated PMN. In vivo experiments showed that circulating PMN-derived microparticles could be detected in mice exposed to bacterial products (LPS/fMLP), being downregulated in animals treated with LMW-Fuc. The data highlight the autocrine and paracrine role of pro-inflammatory microparticles derived from activated PMN and demonstrate the anti-inflammatory effects of LMW-Fuc on these cells.


Asunto(s)
Micropartículas Derivadas de Células/metabolismo , Trampas Extracelulares/metabolismo , Selectina L/metabolismo , Neutrófilos/inmunología , Polisacáridos/farmacología , Animales , Supervivencia Celular , Células Cultivadas , Quimiotaxis , Humanos , Inmunidad Innata , Lipopolisacáridos/inmunología , Ratones , Ratones Endogámicos BALB C , N-Formilmetionina Leucil-Fenilalanina/inmunología , Activación Neutrófila , Estrés Oxidativo , Phaeophyceae/inmunología , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal
18.
Bioimpacts ; 9(1): 25-36, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30788257

RESUMEN

Introduction: The major complications of stent implantation are restenosis and late stent thrombosis. PBMA polymers are used for stent coating because of their mechanical properties. We previously synthesized and characterized Dextrangraft-polybutylmethacrylate copolymer (Dex-PBMA) as a potential stent coating. In this study, we evaluated the haemocompatibility and biocompatibility properties of Dex-PBMA in vitro and in vivo. Methods: Here, we investigated: (1) the effectiveness of polymer coating under physiological conditions and its ability to release Tacrolimus®, (2) the capacity of Dex-PBMA to inhibit Staphylococcus aureus adhesion, (3) the thrombin generation and the human platelet adhesion in static and dynamic conditions, (4) the biocompatibility properties in vitro on human endothelial colony forming cells ( ECFC) and on mesenchymal stem cells (MSC) and in vivo in rat models, and (5) we implanted Dex-PBMA and Dex-PBMATAC coated stents in neointimal hyperplasia restenosis rabbit model. Results: Dex-PBMA coating efficiently prevented bacterial adhesion and release Tacrolimus®. Dex-PBMA exhibit haemocompatibility properties under flow and ECFC and MSC compatibility. In vivo, no pathological foreign body reaction was observed neither after intramuscular nor intravascular aortic implantation. After Dex-PBMA and Dex-PBMATAC coated stents 30 days implantation in a restenosis rabbit model, an endothelial cell coverage was observed and the lumen patency was preserved. Conclusion: Based on our findings, Dex-PBMA exhibited vascular compatibility and can potentially be used as a coating for metallic coronary stents.

19.
Front Med (Lausanne) ; 5: 331, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30538990

RESUMEN

Endothelial progenitor cells (EPCs) are the main hypothetical cells that could give rise to vessels and in particular one subtype isolated from peripheral or cord bloods: endothelial colony forming cells (ECFCs). These ECFCs are clonogenic precursors committed to endothelial lineage and have robust vasculogenic properties. However, their low number and poor expansion properties when isolated from human adult bloods, currently limit their use as an autologous cell therapy product. We previously reported that osteoprotegerin (OPG), a well-characterized regulator of bone metabolism, contributes to ischemic tissue revascularization, tumor growth in vivo, and potentiates ECFCs proangiogenic properties through the secretion of SDF-1. The current study investigated the role of OPG in ECFCs differentiation and expansion from cord blood CD34+ cells. OPG increased the number of ECFCs after endothelial differentiation of CD34+ cells, enhancing the time of EPCs colonies initial appearance and the growth kinetic of endothelial cell progeny. OPG-exposed ECFCs expressed higher levels of CD34+ compared to control ECFCs. In conclusion, our findings provide novel insights into OPG in regulation of CD34+ progenitor cells. These results give new opportunities for ex vivo expansion of human ECFCs using OPG as a cell culture component for future ECFC product manufacture according to GMP.

20.
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
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA