Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
1.
Stem Cells ; 31(9): 1795-805, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23744498

RESUMO

The facilitated recruitment of vascular progenitor cells (VPCs) to ischemic areas might be a therapeutic target for neovascularization and repair. However, efficient and directed attraction of VPCs remains a major challenge in clinical application. To enhance VPC homing, we developed a fusion protein (S1FG), based on the biology of stroma-derived factor-1/CXCL12 and the mucin backbone taken from fractalkine/CXCL12. A GPI-anchor was included to link the fusion-protein to the cell surface. HUVECs transfected with S1FG were capable of increasing firm adhesion of CXCR4+-mononuclear cells (THP-1) under shear stress conditions in vitro. In an in vivo rabbit model of chronic hind limb ischemia, local S1FG application enhanced the recruitment of adoptively transferred embryonic EPCs (eEPCs) to the ischemic muscles 2.5-fold. S1FG combined with eEPC(low) (2 × 10(6)) yielded similar capillary growth as eEPC(high) (5 × 10(6)) alone. Compared to controls, collateral formation was increased in the S1FG eEPC(low) group, but not the eEPC(high) group without S1FG, whereas perfusion was found enhanced in both groups. In addition, S1FG also increased collateral formation and flow when combined with AMD3100 treatment, to increase circulating levels of endogenous VPC. These data demonstrate that the fusion protein S1FG is capable of enhancing the recruitment of exogenously applied or endogenously mobilized progenitor cells to sites of injury. Recombinant versions of S1FG applied via catheters in combination with progenitor cell mobilization may be useful in the treatment of chronic ischemic syndromes requiring improved perfusion.


Assuntos
Quimiocina CX3CL1/metabolismo , Quimiocina CXCL12/metabolismo , Glicosilfosfatidilinositóis/metabolismo , Neovascularização Fisiológica/efeitos dos fármacos , Proteínas Recombinantes de Fusão/farmacologia , Animais , Adesão Celular/efeitos dos fármacos , Endocitose/efeitos dos fármacos , Células Endoteliais da Veia Umbilical Humana/citologia , Células Endoteliais da Veia Umbilical Humana/efeitos dos fármacos , Células Endoteliais da Veia Umbilical Humana/metabolismo , Humanos , Camundongos , Coelhos , Receptores CXCR4/metabolismo , Células-Tronco/citologia , Células-Tronco/efeitos dos fármacos , Células-Tronco/metabolismo
2.
Stem Cells ; 28(2): 376-85, 2010 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-20014279

RESUMO

Embryonal endothelial progenitor cells (eEPCs) are capable of inducing therapeutic angiogenesis in a chronic hind limb model. However, the proportion of eEPCs recruited to the ischemic tissue appears to be a limiting step for the induction of cell-based therapeutic neovascularization. In the present study, we primed eEPCs with the human cathelicidin LL37 (hCAP-18) ex vivo to selectively enhance the eEPC-dependent gain of perfusion in vivo and elucidated the mechanism of action of LL37 on eEPCs. Seven days after femoral artery excision, 5 x 10(6) eEPCs (wt, wild type; p65t, transiently p65 transient; p65s, stable p65-transfected; LL37-eEPCs, LL37 peptide preincubated) were retroinfused into the anterior tibial vein. Recruitment of diI-labeled eEPCs in the ischemic gastrocnemic muscle was investigated 2 days later, whereas collateral growth and perfusion score (obtained by fluorescent microspheres) were assessed at day 7 and day 35 and are given as percentage of day 7 level. Capillary/muscle fiber ratio in the ischemic lower limb was obtained at day 35. Embryonic EPC recruitment in vitro and in vivo was found elevated after LL37 and p65t pretreatment, but not in p65s-eEPCs displaying increased IkappaBalpha or after LL37 in IkappaB-DN overexpressing eEPCs. Using LL37- and p65t-eEPCs, collateral growth (181 +/- 10% and 165 +/- 8%, respectively) surpassed that of wt-eEPCs (135 +/- 7%), increasing perfusion ratio (208 +/- 20% and 210 +/- 17% vs. 142 +/- 12% in wt-eEPCs, respectively), whereas p65s-eEPCs exerted no additive effect (collateral growth 130 +/- 8%; perfusion ratio 155 +/- 15%). Moreover, p65t-eEPC-induced neovascularization was abrogated by blocking antibodies against E-selectin and P-selectin glycoprotein ligand-1 (PSGL-1). We conclude that NF kappaB activation by LL37 or transient p65-transfection increases functionally relevant eEPC recruitment to ischemic muscle tissue via induction of PSGL-1 and E-selectin.


Assuntos
Catelicidinas/farmacologia , Células Endoteliais/citologia , Células Endoteliais/fisiologia , Glicoproteínas de Membrana/metabolismo , Neovascularização Fisiológica/fisiologia , Fragmentos de Peptídeos/farmacologia , Células-Tronco/citologia , Células-Tronco/fisiologia , Fator de Transcrição RelA/fisiologia , Animais , Células Cultivadas , Selectina E/genética , Selectina E/metabolismo , Células Endoteliais/metabolismo , Feminino , Citometria de Fluxo , Humanos , Immunoblotting , Glicoproteínas de Membrana/genética , Neovascularização Fisiológica/efeitos dos fármacos , Neovascularização Fisiológica/genética , Coelhos , Ratos , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Células-Tronco/metabolismo , Fator de Transcrição RelA/genética , Fator de Transcrição RelA/metabolismo
3.
J Clin Invest ; 111(11): 1665-72, 2003 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-12782669

RESUMO

Antimicrobial peptides are effector molecules of the innate immune system and contribute to host defense and regulation of inflammation. The human cathelicidin antimicrobial peptide LL-37/hCAP-18 is expressed in leukocytes and epithelial cells and secreted into wound and airway surface fluid. Here we show that LL-37 induces angiogenesis mediated by formyl peptide receptor-like 1 expressed on endothelial cells. Application of LL-37 resulted in neovascularization in the chorioallantoic membrane assay and in a rabbit model of hind-limb ischemia. The peptide directly activates endothelial cells, resulting in increased proliferation and formation of vessel-like structures in cultivated endothelial cells. Decreased vascularization during wound repair in mice deficient for CRAMP, the murine homologue of LL-37/hCAP-18, shows that cathelicidin-mediated angiogenesis is important for cutaneous wound neovascularization in vivo. Taken together, these findings demonstrate that LL-37/hCAP-18 is a multifunctional antimicrobial peptide with a central role in innate immunity by linking host defense and inflammation with angiogenesis and arteriogenesis.


Assuntos
Antibacterianos/química , Peptídeos Catiônicos Antimicrobianos/farmacologia , Neovascularização Fisiológica , Pró-Fármacos/farmacologia , Receptores de Formil Peptídeo , Receptores de Lipoxinas , Acetilcisteína/farmacologia , Animais , Peptídeos Catiônicos Antimicrobianos/genética , Apoptose , Western Blotting , Cálcio/metabolismo , Catelicidinas , Divisão Celular , Células Cultivadas , Colágeno/farmacologia , Cricetinae , Combinação de Medicamentos , Endotélio Vascular/citologia , Endotélio Vascular/efeitos dos fármacos , Endotélio Vascular/metabolismo , Inibidores Enzimáticos/farmacologia , Humanos , Indóis/farmacologia , Inflamação/metabolismo , Isquemia , Laminina/farmacologia , Maleimidas/farmacologia , Mesocricetus , Camundongos , NF-kappa B/metabolismo , Peptídeos/química , Proteoglicanas/farmacologia , Coelhos , Receptores de Superfície Celular/biossíntese , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fatores de Tempo , Cicatrização
4.
FASEB J ; 19(11): 1576-8, 2005 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-16009705

RESUMO

Clonal embryonic endothelial progenitor cells (eEPCs) isolated from embryonic day 7.5 mice home specifically to hypoxic areas in mouse tumor metastases but spare normal organs and do not form carcinomas. Based on these results, we assessed the potential of eEPCs to enhance vascularization and limit organ dysfunction after ischemia in syngenic and xenotypic organisms. The angiogenic potential of eEPCs was evaluated in chronic ischemic rabbit hindlimbs after regional application by retroinfusion. eEPC treatment improved limb perfusion, paralleled by an increase in capillary density and collateral blood vessel number. Systemic eEPC infusion into mice after ischemic cardiac insult increased postischemic heart output measured by a marked improvement in left ventricle developed pressure and both systolic and diastolic functions. In vitro, eEPCs strongly induced vascular outgrowths from aortic rings. To address the molecular basis of this intrinsic angiogenic potential, we investigated the eEPC transcriptome. Genome-wide Affymetrix GeneChip analysis revealed that the eEPCs express a wealth of secreted factors known to induce angiogenesis, tissue remodeling, and organogenesis that may contribute to the eEPC-mediated beneficial effects. Our findings show that eEPCs induce blood vessel growth and cardioprotection in severe ischemic conditions providing a readily available source to study the mechanisms of neovascularization and tissue recovery.


Assuntos
Embrião de Mamíferos/citologia , Infarto do Miocárdio/terapia , Neovascularização Fisiológica , Transplante de Células-Tronco , Células-Tronco/fisiologia , Animais , Células Cultivadas , Membro Posterior/irrigação sanguínea , Camundongos , Camundongos Endogâmicos C57BL , Traumatismo por Reperfusão Miocárdica/prevenção & controle , Coelhos
5.
Cardiovasc Res ; 65(3): 728-36, 2005 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-15664400

RESUMO

OBJECTIVE: Induction of angiogenesis has been reported subsequent to eNOS overexpression or activation, the latter involving Hsp90 as a chaperone protein. Here, we investigated the potential of regional Hsp90 overexpression to induce therapeutic neovascularization in vivo in a chronic rabbit hindlimb ischemia model. METHODS: In rabbits (n=7 per group), the external femoral artery was excised at day 0 (d0). At d7, liposomes containing eGFP (control group) or Hsp90 were retroinfused into the anterior tibial vein. At day 7 and day 35, angiographies were obtained and analyzed for collateral formation and perfusion velocity (frame count score) (% of d7 values). Capillary/muscle fiber (C/MF) ratio was calculated from five muscle areas of the ischemic limb. L-NAME and Geldanamycin were co-applied, where indicated. RESULTS: Compared to mock-treated controls, Hsp90 transfected increased C/MF ratio at day 35 (1.78+/-0.15 vs. 1.19+/-0.13, p<0.05), an effect blunted by L-NAME (1.39+/-0.11). Hsp90 transfection increased collateral formation (157+/-11% vs. 110+/-13%) and frame count score (174+/-18% vs. 117+/-10%), both sensitive to inhibition by L-NAME coapplication (135+/-17% and 134+/-14%, respectively). Of note, C/MF ratio was found elevated 3 days after Hsp90 transfection (1.61+/-0.16 at d10), at a time point when collateral formation was unchanged (106+/-6%), and tended to remain elevated in the presence of L-NAME applied thereafter (1.64+/-0.35 at d35), though L-NAME blocked subsequent changes in collateral growth or increase in perfusion at d35. CONCLUSIONS: We conclude that Hsp90 is capable of inducing angiogenesis and arteriogenesis via nitric oxide (NO) in a rabbit model of chronic ischemia. Our findings describe the capillary level as an initial site of Hsp90-cDNA-induced neovascularization, followed by growth of larger conductance vessels, resulting in an improved hindlimb perfusion.


Assuntos
Terapia Genética/métodos , Proteínas de Choque Térmico HSP90/genética , Isquemia/terapia , Neovascularização Fisiológica , Óxido Nítrico/fisiologia , Animais , Capilares/patologia , Doença Crônica , DNA Complementar/genética , Feminino , Proteínas de Choque Térmico HSP90/administração & dosagem , Proteínas de Choque Térmico HSP90/biossíntese , Isquemia/metabolismo , Isquemia/patologia , Lipossomos , NG-Nitroarginina Metil Éster/farmacologia , Óxido Nítrico Sintase/metabolismo , Óxido Nítrico Sintase Tipo III , Coelhos , Transfecção
6.
Endothelium ; 10(4-5): 257-65, 2003.
Artigo em Inglês | MEDLINE | ID: mdl-14660086

RESUMO

Therapeutic induction of angiogenesis has been shown in experimental hindlimb ischemia. An alternative to targeting the ischemic hindlimb tissue via the severely stenosed or occluded artery consists in the intact venous system, e.g., by retroinfusion. We tested whether basic fibroblast growth factor (bFGF) enhances angiogenesis induction. Therefore, we applied bFGF retrogradely as compared to intramuscular application. Furthermore, we assessed whether bFGF-induced angiogenesis was enhanced by low-dose VEGF coapplication. Chronic hindlimb ischemia in rabbits was established by excision of the femoral artery at day 0 (d0). At d7, baseline collateral number in the ischemic limb and collateral flow velocity of contrast agent (frame count score) were assessed. Thereafter, saline solution (control group) or bFGF (20 microg/kg) with or without VEGF (10 microg/kg) was retroinfused through the femoral vein. Alternatively, bFGF (20 microg/kg) was injected into thigh and calf muscles. At d35, collateral growth and flow velocity were quantified, and tissue samples were analyzed for capillary density. In the untreated control group, capillary/muscle fiber (C/FM) ratio of the ischemic limb was 0.87 +/- 0.12, and collateral number as well as frame count score at -d35 did not change compared to d7 (107% +/- 7% and 109% +/- 10% of baseline, respectively). Retrograde application of bFGF induced capillary and collateral growth (C/FM ratio 1.56 +/- 0.19 and frame count 161% +/- 29% of baseline), resulting in enhanced flow velocity (143% +/- 13%), similar to the intramuscular application of bFGF. Additional low-dose VEGF retroinfusion did not further increase capillary/collateral growth (1.49 +/- 0.08 and 172% +/- 26%) nor perfusion velocity (149% +/- 7%). The authors conclude that bFGF retroinfusion is a feasible approach of inducing angiogenesis and arteriogenesis in an ischemic hindlimb, resulting in increased blood perfusion, which was not further extended by additional low-dose VEGF coapplication.


Assuntos
Artérias/crescimento & desenvolvimento , Fator 2 de Crescimento de Fibroblastos/uso terapêutico , Isquemia/tratamento farmacológico , Neovascularização Fisiológica/efeitos dos fármacos , Animais , Artérias/efeitos dos fármacos , Velocidade do Fluxo Sanguíneo , Capilares/metabolismo , Capilares/patologia , Doença Crônica , Circulação Colateral , Fator 2 de Crescimento de Fibroblastos/administração & dosagem , Membro Posterior/irrigação sanguínea , Infusões Intravenosas , Injeções Intramusculares , Isquemia/patologia , Isquemia/fisiopatologia , Coelhos , Fator A de Crescimento do Endotélio Vascular/uso terapêutico
7.
Int J Cardiol ; 141(1): e4-7, 2010 May 14.
Artigo em Inglês | MEDLINE | ID: mdl-19131134

RESUMO

We report on a rare case of combined unilateral atresia of the proximal right pulmonary artery (PA) and left patent ductus arteriosus (PDA). A 46 year-old female patient with known PDA and associated advanced pulmonary arterial hypertension presented with progressive hemoptysis and increasing exertional dyspnea. Computed tomography of the chest proved the presence of the known PDA but surprisingly failed to demonstrate an extrapulmonary proximal right PA. We show the imaging features and discuss the embryological background of this rare congenital cardiovascular malformation.


Assuntos
Permeabilidade do Canal Arterial/diagnóstico , Artéria Pulmonar/anormalidades , Artéria Pulmonar/embriologia , Malformações Vasculares/diagnóstico , Fatores Etários , Diagnóstico Diferencial , Permeabilidade do Canal Arterial/complicações , Permeabilidade do Canal Arterial/embriologia , Feminino , Humanos , Pessoa de Meia-Idade , Artéria Pulmonar/diagnóstico por imagem , Tomografia Computadorizada por Raios X , Malformações Vasculares/complicações , Malformações Vasculares/embriologia
8.
J Am Coll Cardiol ; 56(5): 414-22, 2010 Jul 27.
Artigo em Inglês | MEDLINE | ID: mdl-20650363

RESUMO

OBJECTIVES: We set out to investigate the ability of cardiotropic adeno-associated viral vector (AAV2.9 = recombinant adeno-associated virus [rAAV]) to induce prolonged expression of vascular endothelial growth factor (VEGF)-A and platelet-derived growth factor (PDGF)-B in a rabbit hindlimb ischemia model and a pig model of hibernating myocardium. BACKGROUND: Gene therapy to induce angiogenesis and arteriogenesis has produced mixed results. However, long-acting viruses, such as rAAV, as well as combined induction of angiogenesis and vessel maturation might extend the therapeutic potential. METHODS: In rabbits, 0.5 x 10(11) particles rAAV.VEGF-A with or without 1 x 10(12) particles rAAV.PDGF-B were retroinfused at day 7 after femoral artery excision. At days 7 and 35, collateral counts and perfusion were determined, each value given as the day 35/day 7 ratio. Capillary-to-muscle fiber ratio was determined at day 35. In pigs, implantation of a reduction stent graft into the circumflex artery led to complete occlusion at day 28. At this time point, retroinfusion of rAAV.VEGF-A (1 x 10(13) particles), rAAV.VEGF-A/PDGF-B (2 x 10(12) and 4 x 10(12) particles, respectively) or mock transfection was performed. Ejection fraction and left ventricular end-diastolic pressure were assessed at days 28 and 56. RESULTS: In rabbits, rAAV.VEGF-A strongly induced angiogenesis (capillary-to-muscle fiber ratio; 1.67 +/- 0.09 vs. 1.32 +/- 0.11 in rAAV.LacZ-treated limbs, p < 0.05), but not collateral growth (125 +/- 7% vs. 106 +/- 7%, p = NS) or perfusion (136 +/- 12% vs. 107 +/- 9%, p = NS). With VEGF-A/PDGF-B cotransfection, collateral growth increased to 146 +/- 9%, perfusion to 163 +/- 8% of the respective day 7 value (p < 0.05). In the pig model, retroinfusion of rAAV.VEGF-A/PDGF-B increased regional myocardial blood flow reserve from 101 +/- 4% (rAAV.Mock) to 129 +/- 8% (p < 0.05), based on collateral growth (3.2 +/- 0.3 in rAAV.Mock vs. 9.0 +/- 0.4 in rAAV.VEGF-A/PDGF-B, p < 0.05), whereas rAAV.VEGF-A did not alter flow reserve (112 +/- 7%) or collateral count (5.2 +/- 0.7). rAAV.VEGF-A/PDGF-B improved ejection fraction (55 +/- 5% vs. 34 +/- 3% in rAAV.Mock, p < 0.05) unlike rAAV.VEGF-A (37 +/- 2%). CONCLUSIONS: Retroinfusion of rAAV.VEGF-A alone induces angiogenesis, but fails to enhance collateralization and perfusion, unless PDGF-B is cotransfected. In addition to neovascularization, rAAV.VEGF-A/PDGF-B improves regional and global myocardial function in hibernating myocardium.


Assuntos
Vasos Sanguíneos/patologia , Dependovirus/metabolismo , Terapia Genética/métodos , Isquemia/terapia , Miocárdio/patologia , Proteínas Proto-Oncogênicas c-sis/genética , Fator A de Crescimento do Endotélio Vascular/genética , Animais , Técnicas de Transferência de Genes , Técnicas Genéticas , Vetores Genéticos , Neovascularização Fisiológica , Proteínas Proto-Oncogênicas c-sis/uso terapêutico , Coelhos , Suínos , Transfecção , Fator A de Crescimento do Endotélio Vascular/uso terapêutico
9.
Hum Gene Ther ; 20(2): 159-67, 2009 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-20377367

RESUMO

Therapeutic neovascularization is a concept well validated in animal models, however, without clear-cut success in clinical studies. To achieve prolonged transgene expression, recombinant adeno-associated virus (rAAV) was used in a chronic ischemic hind-limb model and the human antimicrobial peptide cathelicidin (LL-37/hCAP-18) was used as proangiogenic factor. Seven days after femoral artery excision, 0.5 x 10(11) rAAV particles encoding for green fluorescent protein (rAAV.GFP), cathelicidin (rAAV.cath), or vascular endothelial growth factor A (rAAV.VEGF-A) were retroinfused into the anterior tibial vein of rabbits (n = 5 per group). In addition, one rAAV.cath-treated group obtained a constant infusion with the phosphatidylinositol 3-kinase (PI3K) inhibitor wortmannin into the ischemic tissue starting on day 7. On day 7 and day 35 angiography of both hind limbs was performed for collateral quantification and frame count score (cinedensitometry). Capillary-to-muscle fiber ratios were obtained on day 35. Compared with controls, application of rAAV.cath induced a gain of perfusion (153 +/- 12 vs. 107 + 9% of day 7 controls) via increased collateral growth (length index, 161 +/- 14 vs. 97 +/- 9%, controls), but no significant capillary growth (1.16 +/- 0.09 vs. 0.99 +/- 0.08, controls). Wortmannin application completely abolished the effects of rAAV.cath, indicating the involvement of the PI3K signal pathway. In conclusion, rAAV-mediated cathelicidin expression is capable of inducing functionally relevant neovascularization, preferentially by collateral growth. The rAAV-based vectors as long-expressing vector expression systems and cathelicidin as proangiogenic factor provide a promising new combination in the treatment of peripheral artery disease.


Assuntos
Catelicidinas/genética , Dependovirus/genética , Isquemia/terapia , Neovascularização Fisiológica/genética , Animais , Peptídeos Catiônicos Antimicrobianos , Catelicidinas/metabolismo , Células Cultivadas , Modelos Animais de Doenças , Técnicas de Transferência de Genes , Membro Posterior/irrigação sanguínea , Humanos , Coelhos , Proteínas Recombinantes/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fator A de Crescimento do Endotélio Vascular/genética , Fator A de Crescimento do Endotélio Vascular/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA