Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
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.
J Thromb Haemost ; 20(1): 230-237, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34623025

RESUMEN

BACKGROUND: The impact of estrogen and testosterone on atherosclerotic cardiovascular disease is well known, but the role of the gonadotropins follicle-stimulating hormone (FSH), luteinizing hormone (LH), and prolactin (PRL) to some extent remain less studied. OBJECTIVES: To explore the angiogenic potential of gonadotropins on endothelial colony-forming cells (ECFCs). METHODS: We examined the effects of various doses of gonadotropins on ECFCs obtained from cord blood by assessing colony number, proliferation, migration, and sprouting ability. Moreover, we studied thrombin generation in ECFCs exposed to gonadotropins by performing a thrombin generation assay. Finally, we determined the levels of circulating gonadotropins in 30 men, to exclude the effect of estrogen, with lower extremity arterial disease (LEAD), in comparison with age- and sex-matched controls. RESULTS: Exposure to FSH, LH, or PRL resulted in an increase in ECFC migration but showed no effect on proliferation or ECFC commitment from cord blood mononuclear cells. Using a three-dimensional fibrin gel assay, we showed that ECFC sprouting was significantly enhanced by gonadotropins. Exposure to FSH also increased the thrombin generation of ECFCs exposed to FSH. Finally, FSH and LH levels in men with LEAD were higher than those in controls. CONCLUSION: Gonadotropins increase ECFC-related angiogenesis and may be involved in thrombin generation in cardiovascular disease. Gonadotropins may act as biomarkers; moreover, we hypothesize that gonadotropin-blocking strategies may be a novel interesting therapeutic approach in atherosclerotic cardiovascular disease.


Asunto(s)
Células Endoteliales , Enfermedades Vasculares , Sangre Fetal , Gonadotropinas , Humanos , Testosterona
3.
Int J Mol Sci ; 22(16)2021 Aug 17.
Artículo en Inglés | MEDLINE | ID: mdl-34445542

RESUMEN

Endoglin (Eng) is an endothelial cell (EC) transmembrane glycoprotein involved in adhesion and angiogenesis. Eng mutations result in vessel abnormalities as observed in hereditary hemorrhagic telangiectasia of type 1. The role of Eng was investigated in endothelial functions and permeability under inflammatory conditions, focusing on the actin dynamic signaling pathway. Endothelial Colony-Forming Cells (ECFC) from human cord blood and mouse lung/aortic EC (MLEC, MAEC) from Eng+/+ and Eng+/- mice were used. ECFC silenced for Eng with Eng-siRNA and ctr-siRNA were used to test tubulogenesis and permeability +/- TNFα and +/- LIM kinase inhibitors (LIMKi). In silico modeling of TNFα-Eng interactions was carried out from PDB IDs 5HZW and 5HZV. Calcium ions (Ca2+) flux was studied by Oregon Green 488 in epifluorescence microscopy. Levels of cofilin phosphorylation and tubulin post-translational modifications were evaluated by Western blot. F-actin and actin-tubulin distribution/co-localization were evaluated in cells by confocal microscopy. Eng silencing in ECFCs resulted in a decrease of cell sprouting by 50 ± 15% (p < 0.05) and an increase in pseudo-tube width (41 ± 4.5%; p < 0.001) compared to control. Upon TNFα stimulation, ECFC Eng-siRNA displayed a significant higher permeability compared to ctr-siRNA (p < 0.01), which is associated to a higher Ca2+ mobilization (p < 0.01). Computational analysis suggested that Eng mitigated TNFα activity. F-actin polymerization was significantly increased in ECFC Eng-siRNA, MAEC+/-, and MLEC+/- compared to controls (p < 0.001, p < 0.01, and p < 0.01, respectively) as well as actin/tubulin distribution (p < 0.01). Furthermore, the inactive form of cofilin (P-cofilin at Ser3) was significantly decreased by 36.7 ± 4.8% in ECFC Eng-siRNA compared to ctr-siRNA (p < 0.001). Interestingly, LIMKi reproduced the absence of Eng on TNFα-induced ECFC-increased permeability. Our data suggest that Eng plays a critical role in the homeostasis regulation of endothelial cells under inflammatory conditions (TNFα), and loss of Eng influences ECFC-related permeability through the LIMK/cofilin/actin rearrangement-signaling pathway.


Asunto(s)
Factores Despolimerizantes de la Actina/metabolismo , Permeabilidad de la Membrana Celular , Endoglina/metabolismo , Células Endoteliales/patología , Inflamación/patología , Quinasas Lim/metabolismo , Neovascularización Patológica/patología , Factores Despolimerizantes de la Actina/genética , Animales , Endoglina/genética , Células Endoteliales/metabolismo , Inflamación/genética , Inflamación/metabolismo , Quinasas Lim/genética , Ratones , Neovascularización Patológica/genética , Neovascularización Patológica/metabolismo
4.
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
5.
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.

6.
Angiogenesis ; 22(2): 325-339, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30607696

RESUMEN

INTRODUCTION: Idiopathic pulmonary fibrosis (IPF) is a devastating disease characterized by obliteration of alveolar architecture, resulting in declining lung function and ultimately death. Pathogenic mechanisms involve a concomitant accumulation of scar tissue together with myofibroblasts activation and a strong abnormal vascular remodeling. Endothelial progenitor cells (ECFC subtype) have been investigated in several human lung diseases as a potential actor in IPF. We previously demonstrated that ECFCs are down-regulated in IPF in contrast to healthy controls. We postulated here that ECFCs might behave as a liquid biopsy in IPF patients and that they exert modified vasculogenic properties. METHODS AND RESULTS: ECFCs isolated from controls and IPF patients expressed markers of the endothelial lineage and did not differ concerning adhesion, migration, and differentiation in vitro and in vivo. However, senescent and apoptotic states were increased in ECFCs from IPF patients as shown by galactosidase staining, p16 expression, and annexin-V staining. Furthermore, conditioned medium of IPF-ECFCs had increased level of interleukin-8 that induced migration of neutrophils in vitro and in vivo. In addition, an infiltration by neutrophils was shown in IPF lung biopsies and we found in a prospective clinical study that a high level of neutrophils in peripheral blood of IPF patients was associated to a poor prognosis. CONCLUSION: To conclude, our study shows that IPF patients have a senescent ECFC phenotype associated with an increased IL-8 secretion potential that might contribute to lung neutrophils invasion during IPF.


Asunto(s)
Células Endoteliales/metabolismo , Células Endoteliales/patología , Fibrosis Pulmonar Idiopática/etiología , Fibrosis Pulmonar Idiopática/patología , Interleucina-8/metabolismo , Células Madre/metabolismo , Células Madre/patología , Adulto , Células Cultivadas , Estudios de Cohortes , Células Endoteliales/fisiología , Estudios de Seguimiento , Francia , Humanos , Fibrosis Pulmonar Idiopática/metabolismo , Fenotipo , Cultivo Primario de Células , Células Madre/fisiología
7.
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.

8.
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
9.
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
10.
Mar Drugs ; 13(12): 7446-62, 2015 Dec 18.
Artículo en Inglés | MEDLINE | ID: mdl-26694425

RESUMEN

Low-molecular-weight fucoidan (LMWF) is a sulfated polysaccharide extracted from brown seaweed that presents antithrombotic and pro-angiogenic properties. However, its mechanism of action is not well-characterized. Here, we studied the effects of LMWF on cell signaling and whole genome expression in human umbilical vein endothelial cells and endothelial colony forming cells. We observed that LMWF and vascular endothelial growth factor had synergistic effects on cell signaling, and more interestingly that LMWF by itself, in the absence of other growth factors, was able to trigger the activation of the PI3K/AKT pathway, which plays a crucial role in angiogenesis and vasculogenesis. We also observed that the effects of LMWF on cell migration were PI3K/AKT-dependent and that LMWF modulated the expression of genes involved at different levels of the neovessel formation process, such as cell migration and cytoskeleton organization, cell mobilization and homing. This provides a better understanding of LMWF's mechanism of action and confirms that it could be an interesting therapeutic approach for vascular repair.


Asunto(s)
Movimiento Celular/efectos de los fármacos , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Neovascularización Fisiológica/efectos de los fármacos , Polisacáridos/farmacología , Citoesqueleto/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Humanos , Peso Molecular , Neovascularización Fisiológica/genética , Fosfatidilinositol 3-Quinasa/metabolismo , Polisacáridos/química , Proteínas Proto-Oncogénicas c-akt/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo
11.
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
12.
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
13.
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
14.
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
15.
Biochem Pharmacol ; 84(8): 1014-23, 2012 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-22796565

RESUMEN

Thrombospondin-1 (TSP-1) gives rise to fragments that have both pro- and anti-angiogenic effects in vitro and in vivo. The TSP-HepI peptide (2.3 kDa), located in the N-terminal domain of TSP-1, has proangiogenic effects on endothelial cells. We have previously shown that TSP-1 itself exhibits a dual effect on endothelial colony-forming cells (ECFC) by enhancing their adhesion through its TSP-HepI fragment while reducing their proliferation and differentiation into vascular tubes (tubulogenesis) in vitro. This effect is likely mediated through CD47 binding to the TSP-1 C-terminal domain. Here we investigated the effect of TSP-HepI peptide on the angiogenic properties of ECFC in vitro and in vivo. TSP-HepI peptide potentiated FGF-2-induced neovascularisation by enhancing ECFC chemotaxis and tubulogenesis in a Matrigel plug assay. ECFC exposure to 20 µg/mL of TSP-HepI peptide for 18 h enhanced cell migration (p < 0.001 versus VEGF exposure), upregulated alpha 6-integrin expression, and enhanced their cell adhesion to activated endothelium under physiological shear stress conditions at levels comparable to those of SDF-1α. The adhesion enhancement appeared to be mediated by the heparan sulfate proteoglycan (HSPG) syndecan-4, as ECFC adhesion was significantly reduced by a syndecan-4-neutralising antibody. ECFC migration and tubulogenesis were stimulated neither by a TSP-HepI peptide with a modified heparin-binding site (S/TSP-HepI) nor when the glycosaminoglycans (GAGs) moieties were removed from the ECFC surface by enzymatic treatment. Ex vivo TSP-HepI priming could potentially serve to enhance the effectiveness of therapeutic neovascularisation with ECFC.


Asunto(s)
Endotelio Vascular/citología , Neovascularización Fisiológica/fisiología , Trombospondina 1/fisiología , Animales , Células Cultivadas , Femenino , Citometría de Flujo , Humanos , Ratones , Trombospondina 1/química
16.
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
17.
Blood ; 111(7): 3442-51, 2008 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-18073349

RESUMEN

The endothelial cell protein C receptor also exists in soluble form in plasma (sEPCR), resulting from ADAM17 cleavage. Elevated sEPCR levels are observed in subjects carrying the A3 haplotype, which is characterized by a Ser219Gly substitution in the transmembrane domain, rendering the receptor more sensitive to cleavage. Because sEPCR production is not completely blocked by metalloprotease inhibition, we looked for another mechanism. Comparing mRNA expression patterns and levels in A3 and non-A3 cells from 32 human umbilical cord veins, we detected a truncated mRNA in addition to the full-length mRNA. This truncated mRNA was 16 times more abundant in A3 human umbilical vein endothelial cells than in non-A3 human umbilical vein endothelial cells and encoded a protein lacking the transmembrane domain. We stably expressed a recombinant form of this protein (rEPCRisoform) and a protein mimicking the plasma sEPCR (rEPCRsol). Functional studies of the purified recombinant proteins revealed that the rEPCRisoform bound to recombinant protein C with similar affinity than rEPCRsol and that it also inhibited the anticoagulant activity of APC. Trace amounts of the EPCR isoform were found in the plasma of A3 subjects. These results suggest that the sEPCRisoform could contribute to the regulatory effect of sEPCR in plasma.


Asunto(s)
Empalme Alternativo/fisiología , Antígenos CD/biosíntesis , Células Endoteliales/metabolismo , ARN Mensajero/metabolismo , Receptores de Superficie Celular/biosíntesis , Venas Umbilicales/metabolismo , Proteínas ADAM/genética , Proteínas ADAM/metabolismo , Proteína ADAM17 , Sustitución de Aminoácidos , Antígenos CD/sangre , Antígenos CD/genética , Células Cultivadas , Células Endoteliales/citología , Receptor de Proteína C Endotelial , Expresión Génica , Haplotipos , Humanos , Mutación Missense , Plasma/metabolismo , Inhibidores de Proteasas/farmacología , Isoformas de Proteínas/biosíntesis , Isoformas de Proteínas/sangre , Isoformas de Proteínas/genética , Estructura Terciaria de Proteína/fisiología , ARN Mensajero/genética , Receptores de Superficie Celular/sangre , Receptores de Superficie Celular/genética , Proteínas Recombinantes/sangre , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Venas Umbilicales/citología
18.
Biochem Pharmacol ; 70(8): 1167-75, 2005 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-16153611

RESUMEN

Endothelial progenitor cell (EPC) transplantation is a potential means of inducing neovascularization in vivo. However, the number of circulating EPC is relatively small, it may thus be necessary to enhance their proangiogenic properties ex vivo prior to injection in vivo. Fucoidan has previously been shown to potentiate in vitro tube formation by mature endothelial cells in the presence of basic fibroblast growth factor (FGF-2). We therefore examined whether fucoidan, alone or combined with FGF-2, could increase EPC proangiogenic potency in vitro. EPC exposure to 10 microg/ml fucoidan induced a proangiogenic phenotype, including cell proliferation (p < 0.01) and migration (p < 0.01); moreover, differentiation into vascular cords occurred in the presence of FGF-2 (p < 0.01). This latter effect correlated with upregulation of the cell-surface #alpha6 integrin subunit of the laminin receptor (p < 0.05). Compared to untreated HUVEC, untreated EPC #alpha6 expression and adhesion to laminin were enhanced two-fold. Fucoidan treatment further enhanced HUVEC but not EPC adhesion to laminin. These results show that fucoidan enhances the proangiogenic properties of EPC and suggest that ex vivo fucoidan preconditioning of EPC might lead to increased neovascularization when injected into ischemic tissues.


Asunto(s)
Endotelio/efectos de los fármacos , Polisacáridos/farmacología , Células Madre/efectos de los fármacos , Proliferación Celular , Separación Celular , Células Cultivadas , Endotelio/citología , Factor 2 de Crecimiento de Fibroblastos/metabolismo , Citometría de Flujo , Humanos , Laminina/metabolismo , Peso Molecular , Neovascularización Fisiológica , Fenotipo , Polisacáridos/química , Células Madre/citología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA