Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Thromb Haemost ; 121(12): 1628-1636, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-33851387

RESUMO

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.


Assuntos
Plaquetas/metabolismo , AMP Cíclico/sangue , Microdomínios da Membrana/metabolismo , Proteínas Associadas à Resistência a Múltiplos Medicamentos/sangue , Agregação Plaquetária , Sistemas do Segundo Mensageiro , Alprostadil/farmacologia , Animais , Plaquetas/efeitos dos fármacos , Microdomínios da Membrana/efeitos dos fármacos , Microdomínios da Membrana/genética , Camundongos Knockout , Proteínas Associadas à Resistência a Múltiplos Medicamentos/genética , Agregação Plaquetária/efeitos dos fármacos , beta-Ciclodextrinas/farmacologia
2.
Arterioscler Thromb Vasc Biol ; 39(10): e219-e232, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-31434496

RESUMO

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.


Assuntos
Apoptose/efeitos dos fármacos , Ceramidas/farmacologia , Células Progenitoras Endoteliais/metabolismo , Neovascularização Fisiológica/efeitos dos fármacos , Regeneração/efeitos dos fármacos , Animais , Diferenciação Celular , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Modelos Animais de Doenças , Células Progenitoras Endoteliais/efeitos dos fármacos , Humanos , Isquemia/tratamento farmacológico , Isquemia/metabolismo , Camundongos , Morfogênese/efeitos dos fármacos , Sensibilidade e Especificidade
3.
Stem Cell Rev Rep ; 15(4): 590-600, 2019 08.
Artigo em Inglês | MEDLINE | ID: mdl-30879244

RESUMO

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.


Assuntos
Antígeno AC133/biossíntese , Antígenos de Diferenciação/biossíntese , Células Progenitoras Endoteliais/metabolismo , Regulação da Expressão Gênica , Neovascularização Fisiológica , Animais , Células Progenitoras Endoteliais/citologia , Xenoenxertos , Membro Posterior/irrigação sanguínea , Células Endoteliais da Veia Umbilical Humana/citologia , Células Endoteliais da Veia Umbilical Humana/metabolismo , Humanos , Isquemia/metabolismo , Isquemia/patologia , Isquemia/terapia , Masculino , Camundongos , Camundongos Nus , Transplante de Células-Tronco
4.
Stem Cell Rev Rep ; 14(6): 812-822, 2018 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-30267203

RESUMO

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.


Assuntos
Bleomicina/efeitos adversos , Células Progenitoras Endoteliais/metabolismo , Pulmão , Fibrose Pulmonar , Microtomografia por Raio-X , Animais , Bleomicina/farmacologia , Modelos Animais de Doenças , Células Progenitoras Endoteliais/patologia , Humanos , Pulmão/diagnóstico por imagem , Pulmão/metabolismo , Camundongos , Camundongos Nus , Fibrose Pulmonar/induzido quimicamente , Fibrose Pulmonar/diagnóstico por imagem , Fibrose Pulmonar/metabolismo
5.
Stem Cell Res Ther ; 9(1): 120, 2018 05 02.
Artigo em Inglês | MEDLINE | ID: mdl-29720269

RESUMO

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.


Assuntos
Ácidos/química , Diabetes Mellitus Experimental/tratamento farmacológico , Células Progenitoras Endoteliais/metabolismo , Glucose/metabolismo , Neovascularização Fisiológica/fisiologia , Animais , Diferenciação Celular , Proliferação de Células , Diabetes Mellitus Experimental/patologia , Modelos Animais de Doenças , Humanos , Masculino , Camundongos , Camundongos Nus
6.
Stem Cell Rev Rep ; 14(1): 82-91, 2018 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-28980146

RESUMO

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.


Assuntos
Fatores de Crescimento Endotelial/metabolismo , Células Progenitoras Endoteliais/citologia , Diferenciação Celular/fisiologia , Movimento Celular/genética , Movimento Celular/fisiologia , Células Cultivadas , Fatores de Crescimento Endotelial/genética , Células Progenitoras Endoteliais/metabolismo , Humanos , MicroRNAs/genética , MicroRNAs/metabolismo , Neovascularização Fisiológica/fisiologia , Interferência de RNA
7.
Thromb Haemost ; 117(10): 1908-1918, 2017 10 05.
Artigo em Inglês | MEDLINE | ID: mdl-28771278

RESUMO

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.


Assuntos
Endoglina/metabolismo , Células Progenitoras Endoteliais/transplante , Isquemia/cirurgia , Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais , Músculo Esquelético/irrigação sanguínea , Neovascularização Fisiológica , Adipogenia , Animais , Adesão Celular , Células Cultivadas , Condrogênese , Modelos Animais de Doenças , Endoglina/genética , Células Progenitoras Endoteliais/metabolismo , Membro Posterior , Isquemia/metabolismo , Isquemia/patologia , Isquemia/fisiopatologia , Masculino , Células-Tronco Mesenquimais/metabolismo , Camundongos Nus , Músculo Esquelético/patologia , Necrose , Fenótipo , Interferência de RNA , Recuperação de Função Fisiológica , Fluxo Sanguíneo Regional , Transdução de Sinais , Fatores de Tempo , Transfecção
8.
Cancer Lett ; 395: 11-19, 2017 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-28263839

RESUMO

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.


Assuntos
Movimento Celular/efeitos dos fármacos , Quimiocina CXCL12/fisiologia , Neoplasias/etiologia , Neovascularização Patológica/induzido quimicamente , Osteoprotegerina/farmacologia , Receptores CXCR4/fisiologia , Animais , Linhagem Celular Tumoral , Fator 2 de Crescimento de Fibroblastos/farmacologia , Humanos , Camundongos , Neoplasias/irrigação sanguínea , Neoplasias/patologia , Ensaios Antitumorais Modelo de Xenoenxerto
9.
Blood ; 126(15): 1823-30, 2015 Oct 08.
Artigo em Inglês | MEDLINE | ID: mdl-26316625

RESUMO

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.


Assuntos
Plaquetas/patologia , AMP Cíclico/metabolismo , Homeostase/fisiologia , Proteínas Associadas à Resistência a Múltiplos Medicamentos/fisiologia , Ativação Plaquetária , Trombose/patologia , Animais , Transporte Biológico , Plaquetas/metabolismo , Células Cultivadas , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Camundongos , Camundongos Knockout , Fosforilação , Agregação Plaquetária , Testes de Função Plaquetária , Transdução de Sinais , Trombose/metabolismo
10.
Int J Cancer ; 137(1): 73-85, 2015 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-25471534

RESUMO

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.


Assuntos
Neoplasias Ósseas/patologia , Interleucinas/metabolismo , Neoplasias Pulmonares/patologia , Macrófagos/metabolismo , Neovascularização Patológica/metabolismo , Osteossarcoma/patologia , Animais , Neoplasias Ósseas/irrigação sanguínea , Neoplasias Ósseas/metabolismo , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Regulação Neoplásica da Expressão Gênica , Células Endoteliais da Veia Umbilical Humana , Humanos , Técnicas In Vitro , Interleucina-1beta/metabolismo , Interleucinas/farmacologia , Neoplasias Pulmonares/irrigação sanguínea , Neoplasias Pulmonares/metabolismo , Camundongos , Transplante de Neoplasias , Osteossarcoma/irrigação sanguínea , Osteossarcoma/metabolismo , Fator de Necrose Tumoral alfa/metabolismo
11.
Angiogenesis ; 17(4): 867-79, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-24854678

RESUMO

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.


Assuntos
Ácidos/química , Meios de Cultura/química , Células Endoteliais/citologia , Células-Tronco/citologia , Animais , Antígenos CD34/metabolismo , Apoptose , Ciclo Celular , Diferenciação Celular , Proliferação de Células , Quimiotaxia , Humanos , Concentração de Íons de Hidrogênio , Isquemia/patologia , Masculino , Camundongos , Camundongos Nus , Neovascularização Fisiológica/efeitos dos fármacos , Cicatrização
12.
Angiogenesis ; 17(3): 603-16, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24419917

RESUMO

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.


Assuntos
Diferenciação Celular , Células Endoteliais/metabolismo , Células-Tronco/metabolismo , Receptor 1 de Fatores de Crescimento do Endotélio Vascular/metabolismo , Animais , Procedimentos Cirúrgicos Cardíacos , Diferenciação Celular/efeitos dos fármacos , Ensaios de Migração Celular , Proliferação de Células/efeitos dos fármacos , Colágeno/metabolismo , Ensaio de Unidades Formadoras de Colônias , Doença da Artéria Coronariana/sangue , Doença da Artéria Coronariana/patologia , Combinação de Medicamentos , Células Endoteliais/efeitos dos fármacos , Membro Posterior/irrigação sanguínea , Membro Posterior/patologia , Humanos , Isquemia/patologia , Laminina/metabolismo , Proteínas de Membrana/sangue , Camundongos Endogâmicos C57BL , Neovascularização Fisiológica/efeitos dos fármacos , Fosforilação/efeitos dos fármacos , Proteoglicanas/metabolismo , RNA Interferente Pequeno/metabolismo , Proteínas Recombinantes/farmacologia , Células-Tronco/efeitos dos fármacos , Fator A de Crescimento do Endotélio Vascular/sangue
13.
Cytotherapy ; 16(2): 213-24, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24280239

RESUMO

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.


Assuntos
Células-Tronco Adultas/metabolismo , Células Endoteliais/metabolismo , Antebraço/fisiopatologia , Membro Posterior/fisiopatologia , Isquemia/fisiopatologia , Adulto , Células-Tronco Adultas/patologia , Idoso , Animais , Diferenciação Celular , Células Cultivadas , Células Endoteliais/patologia , Células Endoteliais/transplante , Feminino , Antebraço/irrigação sanguínea , Membro Posterior/irrigação sanguínea , Humanos , Isquemia/patologia , Masculino , Camundongos , Camundongos Nus , Pessoa de Meia-Idade , Neovascularização Fisiológica , Transplante de Células-Tronco , Células-Tronco , Adulto Jovem
14.
Cardiovasc Res ; 95(1): 39-47, 2012 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-22517984

RESUMO

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.


Assuntos
Integrina alfa6/fisiologia , Isquemia/fisiopatologia , Neovascularização Fisiológica , Receptores Proteína Tirosina Quinases/fisiologia , Animais , Linhagem da Célula , Movimento Celular , Fator 2 de Crescimento de Fibroblastos/farmacologia , Masculino , Camundongos , Camundongos Knockout , Subunidades Proteicas/fisiologia , Receptor TIE-2
15.
Blood ; 118(6): 1699-709, 2011 Aug 11.
Artigo em Inglês | MEDLINE | ID: mdl-21659549

RESUMO

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.


Assuntos
Células Endoteliais/metabolismo , Perfilação da Expressão Gênica , Recém-Nascido de Baixo Peso/sangue , Recém-Nascido Prematuro/sangue , Neovascularização Fisiológica/genética , Células-Tronco/metabolismo , Animais , Vasos Sanguíneos/citologia , Vasos Sanguíneos/crescimento & desenvolvimento , Vasos Sanguíneos/metabolismo , Western Blotting , Proliferação de Células , Células Cultivadas , Células Endoteliais/citologia , Feminino , Sangue Fetal/citologia , Humanos , Recém-Nascido , Masculino , Camundongos , Camundongos Endogâmicos , Camundongos Nus , Fosforilação , Proteínas Proto-Oncogênicas c-akt/genética , Proteínas Proto-Oncogênicas c-akt/metabolismo , Interferência de RNA , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transplante de Células-Tronco/métodos , Células-Tronco/citologia , Trombospondina 1/genética , Trombospondina 1/metabolismo
16.
Arterioscler Thromb Vasc Biol ; 31(3): 551-9, 2011 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21148423

RESUMO

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.).


Assuntos
Proteínas Angiogênicas/sangue , Células Endoteliais/metabolismo , Isquemia/metabolismo , Músculo Esquelético/irrigação sanguínea , Neovascularização Fisiológica , Doença Arterial Periférica/sangue , Células-Tronco/metabolismo , Trombospondina 1/sangue , Proteínas Angiogênicas/administração & dosagem , Proteínas Angiogênicas/deficiência , Proteínas Angiogênicas/genética , Animais , Biomarcadores/sangue , Antígeno CD47/metabolismo , Estudos de Casos e Controles , Adesão Celular , Proliferação de Células , Células Cultivadas , Quimiocina CXCL12/metabolismo , Modelos Animais de Doenças , Células Endoteliais/transplante , Membro Posterior , Humanos , Isquemia/fisiopatologia , Isquemia/cirurgia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Doença Arterial Periférica/fisiopatologia , Doença Arterial Periférica/cirurgia , Fenótipo , Fator de Crescimento Placentário , Proteínas da Gravidez/sangue , Interferência de RNA , Receptores CXCR4/metabolismo , Transplante de Células-Tronco , Trombospondina 1/administração & dosagem , Trombospondina 1/deficiência , Trombospondina 1/genética , Resultado do Tratamento , Fator A de Crescimento do Endotélio Vascular/sangue
17.
Arterioscler Thromb Vasc Biol ; 30(12): 2544-52, 2010 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-20847303

RESUMO

OBJECTIVE: To determine the role of Wnt antagonist Dickkopf (DKK) 1 in human endothelial colony-forming cells (ECFCs) in view of the emerging importance of Wnt pathways in vascular biology. METHODS AND RESULTS: Endothelial progenitor cells have been proposed to be crucial in tumor neovascularization. Recombinant DKK1 has been tested in ECFC angiogenic properties in vitro. DKK1 enhanced ECFC proliferation and the capacity of ECFCs to form pseudotubes in Matrigel. These effects have been attributed to enhancement of vascular endothelial growth factor receptor 2, SDF-1, and CXCR4. DKK1 gene silencing has been realized on ECFCs and mesenchymal stem cells, and we found that DKK1 silencing in the 2 cell types decreased their angiogenic potential. We then examined the possible role of DKK1 in tumor neovasculogenesis and found that blood vessels of breast cancer tissues expressed DKK1 far more strongly in human breast tumors than in normal breast tissues. By studying 62 human breast tumors, we found a significant positive correlation between DKK1 expression and von Willebrand factor. In vivo, DKK1 strongly enhanced the vascularization of Matrigel plugs and increased tumor size in a xenograft model of human breast carcinoma in nude mice. CONCLUSIONS: DKK1 enhances angiogenic properties of ECFCs in vitro and is required for ECFC and mesenchymal stem cell angiogenic phenotypes in vivo. DKK1 also increases tumoral angiogenesis. Thus, we demonstrated a major role of DKK1 in angiogenic processes.


Assuntos
Neoplasias da Mama/metabolismo , Células Endoteliais/metabolismo , Sangue Fetal/citologia , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Neovascularização Patológica/metabolismo , Neovascularização Fisiológica , Células-Tronco/metabolismo , Proteínas Wnt/antagonistas & inibidores , Animais , Neoplasias da Mama/irrigação sanguínea , Neoplasias da Mama/genética , Neoplasias da Mama/patologia , Linhagem Celular Tumoral , Proliferação de Células , Quimiocina CXCL12/metabolismo , Feminino , Humanos , Peptídeos e Proteínas de Sinalização Intercelular/genética , Células-Tronco Mesenquimais/metabolismo , Camundongos , Camundongos Nus , Neovascularização Patológica/genética , Fenótipo , Interferência de RNA , Receptores CXCR4/metabolismo , Proteínas Recombinantes/metabolismo , Transdução de Sinais , Fatores de Tempo , Transfecção , Carga Tumoral , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo , Proteínas Wnt/metabolismo , Fator de von Willebrand/metabolismo
18.
Arterioscler Thromb Vasc Biol ; 30(8): 1569-75, 2010 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-20508204

RESUMO

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.


Assuntos
Células Endoteliais/metabolismo , Integrina alfa6/metabolismo , Isquemia/metabolismo , Músculo Esquelético/irrigação sanguínea , Músculo Esquelético/metabolismo , Neovascularização Fisiológica , Células-Tronco/metabolismo , Animais , Apoptose , Capilares/metabolismo , Capilares/fisiopatologia , Adesão Celular , Movimento Celular , Proliferação de Células , Células Cultivadas , Modelos Animais de Doenças , Células Endoteliais/transplante , Humanos , Integrina alfa6/genética , Isquemia/genética , Isquemia/patologia , Isquemia/fisiopatologia , Fluxometria por Laser-Doppler , Masculino , Camundongos , Camundongos Nus , Músculo Esquelético/patologia , Neovascularização Fisiológica/genética , Fosforilação , Proteínas Proto-Oncogênicas c-akt/metabolismo , Interferência de RNA , Fluxo Sanguíneo Regional , Transplante de Células-Tronco , Transfecção
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA