Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
J Cell Biochem ; 120(6): 10652-10661, 2019 06.
Artigo em Inglês | MEDLINE | ID: mdl-30644609

RESUMO

Vascular endothelial growth factor (VEGF) is a promising molecule for cardiovascular diseases therapy. But lack of a targeted delivery system limits its translation into clinical application. This study aimed to develop stably overexpressing VEGF macrophages for targeted VEGF delivery to injured arteries and determine their potential for repairing of the damaged endothelium. Wire-induced carotid artery injury model was established in atherosclerosis-prone mice. It was observed that the VEGF-modified macrophages were recruited to the site of vascular injury and incorporated into new endothelium formation. VEGF-modified macrophages therapy accelerated reendothelialization and attenuated neointima formation. The VEGF protein level in tissues of injured arteries treated with VEGF-modified macrophages was increased. The upregulated C-C chemokine receptor type 5 (CCR5) and unaltered CCR2 protein levels were verified in VEGF-modified macrophages in vitro. Moreover, enhanced nitric oxide (NO) production in the culture medium of VEGF-modified macrophages was demonstrated. Our results indicated that VEGF-modified macrophages acted as vectors of VEGF targeting injured arteries, promoting the repairing directly by incorporating into new endothelium formation and indirectly by secreting sustainable VEGF and producing NO locally. This study represents a novel therapeutic application of targeted cell therapy with VEGF-modified macrophages for cardiovascular diseases.


Assuntos
Lesões das Artérias Carótidas/metabolismo , Sistemas de Liberação de Medicamentos/métodos , Macrófagos/metabolismo , Neointima/terapia , Fator A de Crescimento do Endotélio Vascular/administração & dosagem , Animais , Aterosclerose/patologia , Lesões das Artérias Carótidas/tratamento farmacológico , Lesões das Artérias Carótidas/patologia , Endotélio Vascular/patologia , Macrófagos/química , Masculino , Camundongos , Camundongos Knockout para ApoE , Neointima/tratamento farmacológico , Neointima/patologia , Óxido Nítrico/metabolismo , Células RAW 264.7 , Receptores CCR2/metabolismo , Receptores CCR5/metabolismo , Fator A de Crescimento do Endotélio Vascular/metabolismo
2.
IUBMB Life ; 71(1): 125-133, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30291803

RESUMO

The long-term failure of vein grafts due to neointimal hyperplasia remains a difficult problem in cardiovascular surgery. Exploring novel approaches to prevent neointimal hyperplasia is important. MicroRNA-146a (miR-146a) plays an essential role in promoting vascular smooth muscle cell (VSMC) proliferation. Thus, the aim of the present study is to investigate whether adenovirus-mediated miR-146a sponge (Ad-miR-146a-SP) gene therapy could attenuate neointimal formation in rat vein grafts. (Ad-miR-146a-SP) was constructed to transfect cultured VSMCs and grafted veins. To improve the efficiency of transferring the miR-146a sponge gene into the grafted veins, 20% poloxamer F-127 gel incorporated with 0.25% trypsin was used to increase adenovirus contact time and penetration. miR-146a-SP transduction significantly reduced the expression of miR-146a both in cultured VSMCs and vein grafts. miR-146a sponge markedly attenuated VSMC proliferation and migration. Consistent with this, miR-146a sponge gene therapy significantly attenuated neointimal formation and also improved blood flow in the vein grafts. Mechanistically, we identified the Krüppel-like factor 4(KLF4) as a potential downstream target gene of miR-146a in vein grafts. Our data show that miR-146a sponge gene therapy could effectively reduce miR-146a activity and attenuate neointimal formation in vein grafts, suggesting its potential therapeutic application for prevention of vein graft failure. © 2018 IUBMB Life, 71(1):125-133, 2019.


Assuntos
Terapia Genética , MicroRNAs/genética , Neointima/terapia , Veias/crescimento & desenvolvimento , Adenoviridae/genética , Animais , Prótese Vascular , Humanos , Fator 4 Semelhante a Kruppel , Fatores de Transcrição Kruppel-Like/genética , Masculino , MicroRNAs/farmacologia , Músculo Liso Vascular/crescimento & desenvolvimento , Músculo Liso Vascular/patologia , Miócitos de Músculo Liso/metabolismo , Neointima/genética , Ratos , Veias/fisiopatologia
3.
J Cell Mol Med ; 23(2): 1528-1540, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-30484954

RESUMO

Intercellular communication between mesenchymal stem cells (MSCs) and their target cells in the perivascular environment is modulated by exosomes derived from MSCs. However, the potential role of exosome-mediated microRNA transfer in neointimal hyperplasia remains to be investigated. To evaluate the effects of MSC-derived exosomes (MSC-Exo) on neointimal hyperplasia, their effects upon vascular smooth muscle cell (VSMC) growth in vitro and neointimal hyperplasia in vivo were assessed in a model of balloon-induced vascular injury. Our results showed that MSC-Exo were internalised by VSMCs and inhibited proliferation and migration in vitro. Further analysis revealed that miR-125b was enriched in MSC-Exo, and repressed the expression of myosin 1E (Myo1e) by targeting its 3' untranslated region. Additionally, MSC-Exo and exosomally transferred miR-125b repressed Myo1e expression and suppressed VSMC proliferation and migration and neointimal hyperplasia in vivo. In summary, our findings revealed that MSC-Exo can transfer miR-125b to VSMCs and inhibit VSMC proliferation and migration in vitro and neointimal hyperplasia in vivo by repressing Myo1e, indicating that miR-125b may be a therapeutic target in the treatment of vascular diseases.


Assuntos
Exossomos/genética , Hiperplasia/genética , MicroRNAs/genética , Miosina Tipo I/genética , Neointima/genética , Lesões do Sistema Vascular/genética , Animais , Apoptose/genética , Movimento Celular/genética , Proliferação de Células/genética , Exossomos/transplante , Regulação da Expressão Gênica no Desenvolvimento , Humanos , Hiperplasia/patologia , Hiperplasia/terapia , Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais/metabolismo , Camundongos , Músculo Liso Vascular/patologia , Miócitos de Músculo Liso , Neointima/patologia , Neointima/terapia , Lesões do Sistema Vascular/patologia , Lesões do Sistema Vascular/terapia
4.
Transl Stroke Res ; 10(4): 413-427, 2019 08.
Artigo em Inglês | MEDLINE | ID: mdl-30191468

RESUMO

Extracellular superoxide dismutase (EC-SOD) has been implicated in regulation of vascular function but its underlying molecular mechanism is largely unknown. These two-step experiments investigate whether hemagglutinating virus of Japan envelope (HVJ-E) vector-mediated EC-SOD gene delivery might protect against neointima formation, vascular inflammation, and reactive oxygen species (ROS) generation, and also explore cell growth signaling pathways. The first in-vitro experiment was performed to assess the transfection efficacy and safety of HVJ-E compared to lipofectamine®. Results revealed that HVJ-E has higher transfection efficiency and lower cytotoxicity than those of lipofectamine®. Another in-vivo study initially used balloon denudation to rat carotid artery, then delivered EC-SOD cDNA through the vector of HVJ-E. Arterial section with H&E staining from the animals 14 days after balloon injury showed a significant reduction of intima-to-media area ratio in EC-SOD transfected arteries when compared with control (empty vector-transfected arteries) (p < 0.05). Arterial tissue with EC-SOD gene delivery also exhibited lower levels of ROS, as assessed by fluorescent microphotography with dihydroethidium staining. Quantitative RT-PCR revealed that EC-SOD gene delivery significantly diminished mRNA expression of tumor necrosis factor (TNF)-α and interleukin (IL)-1ß (p < 0.05 in all comparisons). An immunoblotting assay from vascular smooth muscle cell (VSMC) cultures showed that the EC-SOD transfected group attenuated the activation of MEK1/2, ERK1/2, and Akt signaling significantly. In conclusion, EC-SOD overexpression by HVJ-E vector inhibits neointima hyperplasia, inflammation, and ROS level triggered by balloon injury. The modulation of cell growth-signaling pathways by EC-SOD in VSMCs might play an important role in these inhibitory effects.


Assuntos
Lesões das Artérias Carótidas/terapia , Técnicas de Transferência de Genes , Neointima/terapia , Espécies Reativas de Oxigênio/antagonistas & inibidores , Vírus Sendai , Superóxido Dismutase/administração & dosagem , Proteínas do Envelope Viral/administração & dosagem , Animais , Lesões das Artérias Carótidas/genética , Lesões das Artérias Carótidas/metabolismo , Células Cultivadas , Células HeLa , Humanos , Hiperplasia/genética , Hiperplasia/metabolismo , Hiperplasia/terapia , Inflamação/genética , Inflamação/metabolismo , Inflamação/terapia , Masculino , Neointima/genética , Neointima/metabolismo , Ratos , Ratos Sprague-Dawley , Espécies Reativas de Oxigênio/metabolismo , Vírus Sendai/genética , Superóxido Dismutase/biossíntese , Superóxido Dismutase/genética , Proteínas do Envelope Viral/genética
5.
J Mol Cell Cardiol ; 121: 145-154, 2018 08.
Artigo em Inglês | MEDLINE | ID: mdl-30003882

RESUMO

BACKGROUND: Antiproliferative drugs in drug eluting stents (DES) are associated with complications due to impaired re-endothelialization. Additionally, adventitial neovascularization has been suggested to contribute to in-stent restenosis (ISR). Since Vascular Endothelial Growth Factors (VEGFs) are the key mediators of angiogenesis, we investigated feasibility and efficacy of local gene therapy for ISR utilizing soluble decoy VEGF receptors to reduce biological activity of adventitial VEGFs. METHOD: Sixty-nine adult WHHL rabbit aortas were subjected to endothelial denudation. Six weeks later catheter-mediated local intramural infusion of 1.5x10e10 pfu adenoviruses encoding soluble VEGF Receptor-1 (sVEGFR1), sVEGFR2, sVEGFR3 or control LacZ and bare metal stent implantation were performed in the same aortic segment. Marker protein expression was assessed at 6d in LacZ cohort. Immunohistochemistry, morphometrical analyses and angiography were performed at d14, d42 and d90. RESULTS: Transgene expression was localized to adventitia. All decoy receptors reduced the size of vasa-vasorum at 14d, AdsVEGFR2 animals also had reduced density of adventitial vasa-vasorum, whereas AdsVEGFR3 increased the density of vasa-vasorum. At d42, AdsVEGFR1 and AdsVEGFR2 reduced ISR (15.7 ±â€¯6.9% stenosis, P < 0.01 and 16.5 ±â€¯2.7%, P < 0.05, respectively) vs. controls (28.3 ±â€¯7.6%). Moreover, AdsVEGFR-3 treatment led to a non-significant trend in the reduction of adventitial lymphatics at all time points and these animals had significantly more advanced neointimal atherosclerosis at 14d and 42d vs. control animals. CONCLUSIONS: Targeting adventitial neovascularization using sVEGFR1 and sVEGFR2 is a novel strategy to reduce ISR. The therapeutic effects dissipate at late follow up following short expression profile of adenoviral vectors. However, inhibition of VEGFR3 signaling accelerates neoatherosclerosis.


Assuntos
Constrição Patológica/terapia , Reestenose Coronária/terapia , Terapia Genética , Neointima/terapia , Neovascularização Patológica/tratamento farmacológico , Túnica Adventícia/fisiopatologia , Animais , Aorta/fisiopatologia , Constrição Patológica/genética , Constrição Patológica/fisiopatologia , Reestenose Coronária/genética , Reestenose Coronária/fisiopatologia , Stents Farmacológicos , Endotélio/citologia , Endotélio/efeitos dos fármacos , Endotélio/crescimento & desenvolvimento , Endotélio Vascular/fisiopatologia , Humanos , Neointima/genética , Neointima/fisiopatologia , Neovascularização Patológica/genética , Neovascularização Patológica/fisiopatologia , Coelhos , Vasa Vasorum/fisiopatologia , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/genética , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/uso terapêutico , Fatores de Crescimento do Endotélio Vascular/genética , Fatores de Crescimento do Endotélio Vascular/uso terapêutico
6.
Hum Gene Ther ; 29(7): 816-827, 2018 07.
Artigo em Inglês | MEDLINE | ID: mdl-29382231

RESUMO

Hepatic growth factor (HGF) has been widely used in studies on arterial remodeling after injury, and results turn out to be inconsistent. The changes of endogenous HGF expression after injury also remain controversial. This study clarified the role of exogenous human HGF (hHGF) gene transfer in neointimal hyperplasia and investigated the associated alterations of endogenous HGF and c-Met expressions under endothelial denudation with or without hHGF gene transfer using a balloon-injured rabbit aorta model. Sixty-one rabbits were randomly divided into normal controls, endothelial injury, endothelial injury with hHGF, or the control vector gene transfer groups. On weeks 1, 2, 4, and 8 after injury, neointimal hyperplasia and endothelialization were evaluated by the ratio of neointimal area to medial area (N/M ratio), CD31-positive staining, α-smooth muscle actin, and endothelial nitric oxide synthase expressions using histological analysis, immunohistochemistry staining, or real-time quantitative reverse transcriptase polymerase chain reaction. Endogenous rabbit HGF (rHGF) and c-Met expressions were detected with immunohistochemistry staining and quantitative reverse transcriptase polymerase chain reaction. It was found that expressions of endogeneous rHGF and c-Met in endothelial injury upregulated with peak levels on week 2 or week 4 after injury (p < 0.01). On week 1 after hHGF transfer, neointimal hyperplasia was significantly inhibited (p < 0.001), with decreased α-smooth muscle actin expression (p < 0.05) and improved endothelial cells regeneration and function (p < 0.01). More remarkable overexpression of endogenous rHGF and c-Met mRNAs were detected, and lowered positive staining of rHGF and c-Met was shown in the neointima (p < 0.05). These results demonstrated hHGF gene transfer induced further overexpression of endogenous rHGF and c-Met mRNAs but lowered immunoreactivities of rHGF and c-Met in the neointima, thus leading to significant attenuation of neointimal hyperplasia.


Assuntos
Terapia Genética , Fator de Crescimento de Hepatócito/administração & dosagem , Hiperplasia/terapia , Neointima/terapia , Proteínas Proto-Oncogênicas c-met/genética , Actinas/genética , Animais , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/metabolismo , Regulação da Expressão Gênica/genética , Vetores Genéticos/administração & dosagem , Fator de Crescimento de Hepatócito/genética , Humanos , Hiperplasia/genética , Hiperplasia/patologia , Neointima/genética , Neointima/patologia , RNA Mensageiro/genética , Coelhos
7.
Exp Toxicol Pathol ; 69(8): 598-604, 2017 Oct 02.
Artigo em Inglês | MEDLINE | ID: mdl-28583699

RESUMO

Although still a matter of controversial discussion, skeletal myoblasts are one of the options for stem cell transplantation improving cardiac function after myocardial infarction, exhibiting several advantages including the availability, the ability of self-renewal and differentiation, and the lack of ethical and immunological problems. The aim of this study was to investigate the impact of stem cell therapy with skeletal myoblasts on experimental venous bypass grafts in a mouse model of vein graft disease. Forty C57BL/6J mice underwent bypass grafting interposing a venous bypass graft of the donor mouse into the carotid artery of the recipient mouse. Twenty mice received periadventitially treatment with 1 million fluorescence labeled skeletal myoblasts suspended in culture medium (treatment group), the other twenty mice received only culture medium without myoblasts (control group). Two weeks after bypass surgery, the vein grafts of all 40 mice were harvested, stained and histologically investigated under light and immunofluorescence microscope. Against our expectations, skeletal myoblasts stayed in place and were still located in the adventitia after bypass grafting. Additionally, vein grafts of the myoblast group revealed a 2fold increased neoneointima formation, a decreased media thickness, a slightly increased neovascularization, a higher percentage of reendothelialization and also a slightly higher percentage of PDGFR ɑ, PDGFR ß, MMP-7 and MMP-9 positive cells, suggesting a paracrine mechanism responsible for accelerated neointima formation. In conclusion, the results of our study do not support the use of skeletal myoblast for the treatment of vein graft disease after coronary artery bypass surgery.


Assuntos
Oclusão de Enxerto Vascular/prevenção & controle , Mioblastos Esqueléticos/transplante , Neointima/terapia , Transplante de Células-Tronco , Animais , Artéria Carótida Primitiva/cirurgia , Ponte de Artéria Coronária , Oclusão de Enxerto Vascular/patologia , Camundongos Endogâmicos C57BL , Resultado do Tratamento , Veia Cava Inferior/transplante
8.
J Cardiol ; 70(3): 297-302, 2017 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-28034576

RESUMO

BACKGROUND: First-generation drug-eluting stents (DES) have reduced short-term stent failure as compared to bare-metal stents due to the inhibition of neointima hyperplasia, but instead increased the risk of very-late stent failure. Although better outcomes have been reported for second-generation DES than for first-generation DES, the difference in the angioscopic findings at 1-year follow-up has not been adequately elucidated among second-generation DES. METHODS: Consecutive 161 patients who received angioscopic examination at 1 year after implantation of second-generation DES, i.e. Nobori biolimus-eluting stents (Terumo, Tokyo, Japan) (N-BES, n=25), Xience everolimus-eluting stents (Abbot Vascular, Santa Clara, CA, USA; X-EES, n=95), or Resolute zotarolimus-eluting stents (Resolute Integrity; Medtronic, Minneapolis, MN, USA; R-ZES, n=41), in de novo native coronary lesions were analyzed. RESULTS: Maximum neointima coverage grade (N-BES, 0.9±0.3; X-EES, 1.2±0.4; R-ZES, 1.5±0.5; p<0.001) was the highest in R-ZES and lowest in N-BES. Heterogeneity score was higher in R-ZES than in N-BES (N-BES, 0.8±0.4; X-EES, 0.9±0.4; R-ZES, 1.1±0.5; p=0.007). Maximum yellow color grade and prevalence of thrombus were not different. Multivariate analysis demonstrated that only stent type was associated with maximum neointima coverage grade; stent type and total stent length were associated with heterogeneity score; and stenting for acute coronary syndrome (ACS) and total stent length were associated with maximum yellow color grade. CONCLUSIONS: Neointima coverage and heterogeneity were mainly determined by stent type even among second-generation DES, while yellow color was determined mainly by whether target lesion was of ACS.


Assuntos
Angioscopia , Stents Farmacológicos , Neointima/diagnóstico por imagem , Síndrome Coronariana Aguda/diagnóstico por imagem , Síndrome Coronariana Aguda/patologia , Síndrome Coronariana Aguda/terapia , Idoso , Idoso de 80 Anos ou mais , Everolimo/administração & dosagem , Feminino , Seguimentos , Humanos , Masculino , Pessoa de Meia-Idade , Neointima/patologia , Neointima/terapia , Sirolimo/administração & dosagem , Sirolimo/análogos & derivados , Trombose/diagnóstico por imagem , Trombose/etiologia , Trombose/patologia , Resultado do Tratamento
9.
Biosci Rep ; 36(6)2016 12.
Artigo em Inglês | MEDLINE | ID: mdl-27815507

RESUMO

Coronary artery disease is treated by vein grafting and stent implantation. Late vein graft failure and restenosis of stented arteries reduce the success rates of these approaches and are caused by neointima formation. We have previously shown that Wnt proteins are up-regulated during intimal thickening, and have speculated that these lead to activation of downstream genes with ß-catenin/T-cell factor (TCF)-responsive promoters. In the present study, we aimed to provide evidence that ß-catenin/TCF signalling promotes neointima formation and assess whether targeting this pathway has potential for reducing neointima formation. We utilized a gene therapy approach selectively targeting cells in which the ß-catenin/TCF pathway is activated by using a recombinant adenovirus Ad-TOPTK, which carries a herpes simplex virus thymidine kinase (HSV-TK) gene under the control of a ß-catenin/TCF-response promoter. Cells with activated ß-catenin will therefore be selectively killed. Ad-TOPTK and ganciclovir (GCV) treatment significantly suppressed the growth of the neointima in a murine model of left carotid artery ligation. In summary, we demonstrated that Wnt/ß-catenin/TCF signalling promotes neointima formation, by showing that the selective death of cells with activated ß-catenin suppressed neointima formation. This highlights the therapeutic potential for reducing late vein graft failure and in-stent restenosis by targeting ß-catenin/TCF signalling.


Assuntos
Neointima/metabolismo , Neointima/terapia , Fatores de Transcrição TCF/metabolismo , beta Catenina/metabolismo , Adenoviridae/genética , Animais , Antivirais/farmacologia , Células Cultivadas , Ganciclovir/farmacologia , Vetores Genéticos/genética , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Neointima/genética , Regiões Promotoras Genéticas/efeitos dos fármacos , Regiões Promotoras Genéticas/genética , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética , Simplexvirus/genética , Timidina Quinase/genética , Regulação para Cima/efeitos dos fármacos , Regulação para Cima/genética , Via de Sinalização Wnt/efeitos dos fármacos , Via de Sinalização Wnt/genética
10.
Int J Cardiol ; 208: 79-86, 2016 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-26828387

RESUMO

BACKGROUND: Vein graft failure due to neointimal hyperplasia remains an important and unresolved problem of cardiovascular surgery. MicroRNA-221 (miR-221) has been shown to play a major role in regulating vascular smooth muscle cell (VSMC) proliferation and phenotype transformation. Thus, the purpose of this study is to determine whether adenovirus mediated miR-221 sponge gene therapy could inhibit vein graft neointimal hyperplasia. METHODS: Adenovirus encoding miR-221 sponge (Ad-miR-221-SP) was used to inhibit VSMC proliferation in vitro and neointimal formation in vivo. Expression of miRNA-221 was evaluated in cultured VSMC and in rat vein graft models following transduction with Ad-miR-221-SP, Ad-Control-SP (without miR-221 antisense binding sites), or Ad-GFP (control). To accelerate the transfer of miR-221 sponge gene to the vein grafts, 20% poloxamer F-127 gel was used to extend virus contact time and 0.25% trypsin to increase virus penetration. RESULTS: miR-221 sponges can significantly decrease the expression of miR-221 and proliferation in cultured VSMC. Cellular proliferation rates were significantly reduced in miR-221 sponge treated grafts as compared with controls at 6 weeks after bypass surgery (19.8% versus 43.6%, P=0.0028). miR-221 sponge gene transfer reduced the neointimal area (210.75 ± 24.13 versus 67.01 ± 12.02, P<0.0001), neointimal thickness (171.86 ± 27.87 versus 64.13 ± 16.23, P<0.0001) and neointima/media ratio (0.74 ± 0.21 versus 1.95 ± 0.25, P<0.0001) in vein grafts versus controls. miR-21 sponge treatment was also improved hemodynamics in vein grafts. We have further identified that p27 (Kip1) is a potential target gene of miR-221 in vein grafts. CONCLUSION: miR-221 sponge therapy can significantly reduce miR-221 activity and inhibit neointimal hyperplasia in vein grafts. Locally adventitial delivery of adenoviruses mediated miRNA sponges may be promising gene therapies to prevent vein graft failure.


Assuntos
Velocidade do Fluxo Sanguíneo/fisiologia , Terapia Genética/métodos , Veias Jugulares/transplante , MicroRNAs/administração & dosagem , Neointima/terapia , Enxerto Vascular/métodos , Adenoviridae/genética , Animais , Células Cultivadas , Hiperplasia/genética , Hiperplasia/fisiopatologia , Hiperplasia/terapia , Veias Jugulares/fisiologia , Masculino , MicroRNAs/genética , Músculo Liso Vascular/fisiologia , Músculo Liso Vascular/transplante , Neointima/genética , Neointima/fisiopatologia , Ratos , Ratos Sprague-Dawley
11.
Mol Ther ; 24(4): 779-87, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26732878

RESUMO

Inhibition of vascular smooth muscle cell (VSMC) proliferation by drug eluting stents has markedly reduced intimal hyperplasia and subsequent in-stent restenosis. However, the effects of antiproliferative drugs on endothelial cells (EC) contribute to delayed re-endothelialization and late stent thrombosis. Cell-targeted therapies to inhibit VSMC remodeling while maintaining EC health are necessary to allow vascular healing while preventing restenosis. We describe an RNA aptamer (Apt 14) that functions as a smart drug by preferentially targeting VSMCs as compared to ECs and other myocytes. Furthermore, Apt 14 inhibits phosphatidylinositol 3-kinase/protein kinase-B (PI3K/Akt) and VSMC migration in response to multiple agonists by a mechanism that involves inhibition of platelet-derived growth factor receptor (PDGFR)-ß phosphorylation. In a murine model of carotid injury, treatment of vessels with Apt 14 reduces neointimal formation to levels similar to those observed with paclitaxel. Importantly, we confirm that Apt 14 cross-reacts with rodent and human VSMCs, exhibits a half-life of ~300 hours in human serum, and does not elicit immune activation of human peripheral blood mononuclear cells. We describe a VSMC-targeted RNA aptamer that blocks cell migration and inhibits intimal formation. These findings provide the foundation for the translation of cell-targeted RNA therapeutics to vascular disease.


Assuntos
Aptâmeros de Nucleotídeos/farmacologia , Músculo Liso Vascular/citologia , Miócitos de Músculo Liso/efeitos dos fármacos , Neointima/terapia , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Animais , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Regulação da Expressão Gênica/efeitos dos fármacos , Meia-Vida , Humanos , Camundongos , Músculo Liso Vascular/efeitos dos fármacos , Miócitos de Músculo Liso/citologia , Neointima/metabolismo , Fosforilação , Ratos
12.
J Mol Cell Cardiol ; 86: 121-35, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26231083

RESUMO

AIMS: It is still controversial whether bone marrow (BM)-derived endothelial progenitor cells (EPCs) can contribute to vascular repair and prevent the progression of vascular diseases. We aimed to characterize BM-derived EPC subpopulations and to evaluate their therapeutic efficacies to repair injured vascular endothelium of systemic and pulmonary arteries. METHODS AND RESULTS: BM mononuclear cells of Fisher-344 rats were cultured under endothelial cell-conditions. Early EPCs appeared on days 3-6. Late-outgrowth and very late-outgrowth EPCs (LOCs and VLOCs) were defined as cells forming cobblestone colonies on days 9-14 and 17-21, respectively. Among EPC subpopulations, LOCs showed the highest angiogenic capability with enhanced proliferation potential and secretion of proangiogenic proteins. To investigate the therapeutic effects of these EPCs, Fisher-344 rats underwent wire-mediated endovascular injury in femoral artery (FA) and were concurrently injected intraperitoneally with 60mg/kg monocrotaline (MCT). Injured rats were then treated with six injections of one of three EPCs (1×10(6) per time). After 4weeks, transplanted LOCs, but not early EPCs or VLOCs, significantly attenuated neointimal lesion formation in injured FAs. Some of CD31(+) LOCs directly replaced the injured FA endothelium (replacement ratio: 11.7±7.0%). In contrast, any EPC treatment could neither replace MCT-injured endothelium of pulmonary arterioles nor prevent the progression of pulmonary arterial hypertension (PAH). LOCs modified protectively the expression profile of angiogenic and inflammatory genes in injured FAs, but not in MCT-injured lungs. CONCLUSION: BM-derived LOCs can contribute to vascular repair of injured systemic artery; however, even they cannot rescue injured pulmonary vasculature under MCT-induced PAH.


Assuntos
Células Progenitoras Endoteliais/transplante , Endotélio Vascular/crescimento & desenvolvimento , Hipertensão Pulmonar/patologia , Neointima/patologia , Doenças Vasculares/patologia , Animais , Arteríolas/crescimento & desenvolvimento , Arteríolas/transplante , Células da Medula Óssea/patologia , Transplante de Medula Óssea , Diferenciação Celular/genética , Proliferação de Células , Células Progenitoras Endoteliais/efeitos dos fármacos , Células Progenitoras Endoteliais/metabolismo , Endotélio Vascular/patologia , Artéria Femoral/efeitos dos fármacos , Artéria Femoral/lesões , Artéria Femoral/patologia , Humanos , Hipertensão Pulmonar/terapia , Monocrotalina/administração & dosagem , Neointima/terapia , Ratos , Doenças Vasculares/terapia
13.
Gene Ther ; 20(12): 1158-64, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24005577

RESUMO

Ex vivo gene therapy during coronary artery bypass grafting (CABG) holds great potential to prevent excessive smooth muscle cell (SMC) proliferation, neointima formation and graft failure. The most successful preclinical strategies to date have utilised vectors based on the species C adenovirus, Ad5, which engages the Coxsackie and Adenovirus receptor (CAR) as its primary attachment receptor. Profiling receptors on human SMCs demonstrated the absence of CAR but substantial expression of the species B receptor CD46. We performed transduction experiments using Ad5 and the CD46-utilising adenovirus Ad35, and found Ad35 significantly more efficient at transducing SMCs. To evaluate whether transduction could be further augmented, we evaluated chimeric CD46-utilising Ad5/Ad35 vectors comprising the Ad5 capsid pseudotyped with the Ad35 fibre alone (Ad5/F35) or in combination with the Ad35 penton (Ad5/F35/P35). In human smooth muscle cells (hSMCs), Ad5/F35/P35 mediated significantly higher levels of transduction than either parental vector or Ad5/F35. Ex vivo transduction experiments using mouse aortas from CD46 transgenics demonstrated that Ad5/F35/P35 was significantly more efficient at transducing SMCs than the other vectors tested. Finally, ex vivo transduction and immunofluorescent colocalisation experiments using human tissue from CABG procedures confirmed the preclinical potential of Ad5/F35/P35 as an efficient vector for vascular transduction during CABG.


Assuntos
Adenovírus Humanos/genética , Proteínas do Capsídeo/metabolismo , Capsídeo/metabolismo , Músculo Liso Vascular/citologia , Músculo Liso Vascular/metabolismo , Transdução Genética , Adenovírus Humanos/classificação , Animais , Aorta , Células Cultivadas , Ponte de Artéria Coronária , Doença da Artéria Coronariana/complicações , Doença da Artéria Coronariana/patologia , Doença da Artéria Coronariana/cirurgia , Doença da Artéria Coronariana/terapia , Proteína de Membrana Semelhante a Receptor de Coxsackie e Adenovirus/metabolismo , Terapia Genética , Vetores Genéticos , Humanos , Proteína Cofatora de Membrana/genética , Proteína Cofatora de Membrana/metabolismo , Camundongos , Camundongos Transgênicos , Músculo Liso Vascular/virologia , Neointima/patologia , Neointima/terapia
14.
Arterioscler Thromb Vasc Biol ; 33(5): 1036-45, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-23430616

RESUMO

OBJECTIVE: Atherosclerosis and restenosis are multifactorial diseases associated with abnormal vascular smooth muscle cell (VSMC) proliferation. Nuclear factor-Y (NF-Y) plays a major role in transcriptional activation of the CYCLIN B1 gene (CCNB1), a key positive regulator of cell proliferation and neointimal thickening. Here, we investigated the role of NF-Y in occlusive vascular disease. APPROACH AND RESULTS: We performed molecular and expression studies in cultured cells, animal models, and human tissues. We find upregulation of NF-Y and cyclin B1 expression in proliferative regions of murine atherosclerotic plaques and mechanically induced lesions, which correlates with higher binding of NF-Y to target sequences in the CCNB1 promoter. NF-YA expression in neointimal lesions is detected in VSMCs, macrophages, and endothelial cells. Platelet-derived growth factor-BB, a main inductor of VSMC growth and neointima development, induces the recruitment of NF-Y to the CCNB1 promoter and augments both CCNB1 mRNA expression and cell proliferation through extracellular signal-regulated kinase 1/2 and Akt activation in rat and human VSMCs. Moreover, adenovirus-mediated overexpression of a NF-YA-dominant negative mutant inhibits platelet-derived growth factor-BB-induced CCNB1 expression and VSMC proliferation in vitro and neointimal lesion formation in a mouse model of femoral artery injury. We also detect NF-Y expression and DNA-binding activity in human neointimal lesions. CONCLUSIONS: Our results identify NF-Y as a key downstream effector of the platelet-derived growth factor-BB-dependent mitogenic pathway that is activated in experimental and human vasculoproliferative diseases. They also identify NF-Y inhibition as a novel and attractive strategy for the local treatment of neointimal formation induced by vessel denudation.


Assuntos
Fator de Ligação a CCAAT/fisiologia , Músculo Liso Vascular/citologia , Neointima/etiologia , Animais , Apolipoproteínas E/fisiologia , Aterosclerose/etiologia , Becaplermina , Fator de Ligação a CCAAT/antagonistas & inibidores , Proliferação de Células , Células Cultivadas , Ciclina B1/genética , Células Endoteliais/fisiologia , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Neointima/terapia , Proteínas Proto-Oncogênicas c-sis/farmacologia , Ratos , Ratos Wistar
15.
J Cardiothorac Surg ; 7: 57, 2012 Jun 20.
Artigo em Inglês | MEDLINE | ID: mdl-22716287

RESUMO

BACKGROUND: Neointimal hyperplasia involving smooth muscle cell (SMC) proliferation, migration and extracellular matrix (ECM) degradation is an important component of atherosclerosis. It develops as a response to vascular injury after balloon angioplasty and vascular graft placement. Matrix metalloproteinases (MMPs) induce SMC proliferation, migration and contribute to intimal hyperplasia by degrading ECM. PPARγ agonists inhibit SMC proliferation, migration and lesion formation. In this study, we aimed to investigate the effects of PPARγ agonist rosiglitazone on neointimal hyperplasia and gelatinase (MMP-2 and MMP-9) expressions in rabbit carotid anastomosis model. METHODS: New Zealand white rabbits (n = 13, 2.7-3.2 kg) were divided into placebo and treatment groups. Right carotid artery (CA) was transected and both ends were anastomosed. Treatment group (n = 6) received rosiglitazone (3 mg/kg/day/p.o.) and placebo group (n = 7) received PBS (phosphate buffered saline, 2.5 ml/kg/day/p.o.) for 4 weeks postoperatively. After the sacrification, right and left CAs were isolated. Morphometric analyses and immunohistochemical examinations for gelatinases were performed. RESULTS: Intimal area (0.055 ± 0.005 control vs 0.291 ± 0.020 µm(2) anastomosed, p < 0,05) and index (0.117 ± 0.002 control vs 0.574 ± 0.013 anastomosed, p < 0,01) significantly increased in anastomosed arteries compared to control arteries from placebo group. However, in rosiglitazone-treated group, intimal area (0.291 ± 0.020 PBS vs 0.143 ± 0.027 rosiglitazone, p < 0,05) and index (0.574 ± 0.013 PBS vs 0.263 ± 0.0078 rosiglitazone, p < 0,01) significantly decreased. Furthermore, gelatinase immunopositivity was found to have significantly increased in anastomosed arteries from placebo group and decreased with rosiglitazone treatment. CONCLUSIONS: These results suggest that rosiglitazone may prevent neointimal hyperplasia, which is the most important factor involved in late graft failure, by inhibiting gelatinase enzyme expression.


Assuntos
Anastomose Cirúrgica/métodos , Artérias Carótidas/cirurgia , Neointima/patologia , Neointima/terapia , PPAR gama/agonistas , Tiazolidinedionas/farmacologia , Análise de Variância , Animais , Procedimentos Cirúrgicos Cardíacos , Artérias Carótidas/efeitos dos fármacos , Artérias Carótidas/patologia , Distribuição de Qui-Quadrado , Feminino , Hiperplasia , Imuno-Histoquímica , Masculino , Metaloproteinases da Matriz/metabolismo , Neointima/tratamento farmacológico , Neointima/prevenção & controle , Coelhos , Rosiglitazona , Estatísticas não Paramétricas
16.
Circulation ; 124(11 Suppl): S135-42, 2011 Sep 13.
Artigo em Inglês | MEDLINE | ID: mdl-21911803

RESUMO

BACKGROUND: Coronary artery vein graft failure, resulting from thrombosis, intimal thickening, and atherosclerosis, is a significant clinical problem, with approximately 50% of vein grafts failing within 10 years. Intimal thickening is caused by migration of vascular smooth muscle cells from the media to the intima, where they proliferate. Interventions using gene transfer to inhibit vascular smooth muscle cells proliferation and migration are attractive because ex vivo access to the graft is possible. The involvement of matrix-degrading metalloproteinases in intimal thickening is well established, and we previously showed that adenoviral-delivered overexpression of an endogenous inhibitor, the tissue inhibitor of metalloproteinases-3 (TIMP-3), significantly retarded intimal thickening in short-term autologous porcine arteriovenous interposition grafts (28 days). However, it is essential to determine whether this approach will provide longer-term benefits. METHODS AND RESULTS: We assessed whether a recombinant adenovirus that overexpresses TIMP-3 (RAdTIMP-3) affects vein graft intimal thickening in the longer term (at 3 months). Porcine saphenous veins were subjected to luminal infection with 2.5×10(10) pfu/mL RAdTIMP-3 or RAd60 (control virus) or vehicle control, for 30 minutes before implantation into the carotid artery. Analysis of grafts harvested 3 months after delivery revealed that RAdTIMP-3-infected grafts had significantly reduced intimal areas compared with both controls (3.2 ± 0.4 mm(2) versus 5.6 ± 0.7 mm(2) and 5.9 ± 0.5 mm(2), RAdTIMP-3, RAd60, and vehicle, respectively). Medial areas were also significantly decreased by TIMP-3 (3.8 ± 0.3 mm(2) versus 6.7 ± 1.0 mm(2) and 5.2 ± 0.4 mm(2), RAdTIMP-3, RAd60, and vehicle, respectively). CONCLUSIONS: Overexpression of TIMP-3 provides a sustained retardation of vein graft intimal thickening and highlights the translational potential for ex vivo TIMP-3 gene therapy.


Assuntos
Artérias Carótidas/cirurgia , Terapia Genética/métodos , Neointima/prevenção & controle , Neointima/terapia , Veia Safena/transplante , Inibidor Tecidual de Metaloproteinase-3/genética , Enxerto Vascular/métodos , Adenoviridae/genética , Animais , Artérias Carótidas/metabolismo , Artérias Carótidas/patologia , Movimento Celular/fisiologia , Endotélio Vascular/metabolismo , Endotélio Vascular/patologia , Modelos Animais , Músculo Liso Vascular/metabolismo , Músculo Liso Vascular/patologia , Veia Safena/metabolismo , Veia Safena/patologia , Suínos , Fatores de Tempo , Inibidor Tecidual de Metaloproteinase-3/metabolismo , Resultado do Tratamento
17.
Cytotherapy ; 13(6): 695-704, 2011 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-21250866

RESUMO

BACKGROUND AIMS: Previously, we have demonstrated that administration of dermatan sulfate (DS) suppresses neointima formation in the mouse carotid artery by activating heparin co-factor II. A similar suppressive effect was observed by increasing the number of progenitor cells in circulation. In this study, we investigated the combination of DS and bone marrow mononuclear cells (MNC), which includes potential endothelial progenitors, in neointima formation after arterial injury. METHODS: Arterial injury was induced by mechanical dilation of the left common carotid artery. We analyzed the extension of endothelial lesion, thrombus formation, P-selectin expression and CD45(+) cell accumulation 1 and 3 days post-injury, and neointima formation 21 days post-injury. Animals were injected with MNC with or without DS during the first 48 h after injury. RESULTS: The extension of endothelial lesion was similar in all groups 1 day after surgery; however, in injured animals treated with MNC and DS the endothelium recovery seemed to be more efficient 21 days after lesion. Treatment with DS inhibited thrombosis, decreased CD45(+) cell accumulation and P-selectin expression at the site of injury, and reduced the neointimal area by 56%. Treatment with MNC reduced the neointimal area by 54%. The combination of DS and MNC reduced neointima formation by more than 91%. In addition, DS promoted a greater accumulation of MNC at the site of injury. CONCLUSIONS: DS inhibits the initial thrombotic and inflammatory processes after arterial injury and promotes migration of MNC to the site of the lesion, where they may assist in the recovery of the injured endothelium.


Assuntos
Células da Medula Óssea/citologia , Artérias Carótidas/efeitos dos fármacos , Dermatan Sulfato/uso terapêutico , Neointima/prevenção & controle , Neointima/terapia , Animais , Anticoagulantes/uso terapêutico , Células da Medula Óssea/fisiologia , Artérias Carótidas/metabolismo , Antígenos Comuns de Leucócito/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Selectina-P/metabolismo , Trombose/prevenção & controle , Trombose/terapia
18.
Chin Med J (Engl) ; 124(24): 4286-92, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-22340401

RESUMO

BACKGROUND: Recent studies indicate that bone marrow-derived cells may significantly contribute to atherosclerosis, post-angioplasty restenosis and transplantation-associated vasculopathy. The responsible bone marrow (BM) cells and mechanisms regulating the mobilization of these cells are currently unclear. The purpose of this study was to investigate the expression of granulocyte colony-stimulating factor (G-CSF) on injured arteries and its effects on mesenchymal stem cells (MSCs) differentiation into vascular smooth muscle cells (VSMCs) in the process of vascular remodeling. METHODS: Balloon-mediated vascular injury was established in female rats (n = 100) which received radioprotective whole female BM cells by tail vein injection and male MSCs through a tibial BM injection after lethal irradiation. The injured and contralateral carotid arteries were harvested at 3, 7, 14 and 28 days after treatment. RESULTS: Morphometric analysis indicated that intima to media area-ratio (I/M ratio) significantly increased at 28 days, 0.899 ± 0.057 (P < 0.01), compared with uninjured arteries. Combining fluorescence in situ hybridization (FISH) and immunohistochemical analysis showed that a significant number of the neointimal cells derived from MSCs, (45.2 ± 8.5)% at 28 days (P = 0.01), compared with (23.5 ± 6.3)% at 14 days. G-CSF was induced in carotid arteries subject to balloon angioplasty (fold mRNA change = 8.67 ± 0.63 at three days, relative G-CSF protein = 0.657 ± 0.011 at three days, P < 0.01, respectively, compared with uninjured arteries). G-CSF was chemotactic for MSCs but did not affect the differentiation of MSCs into smooth-muscle-like cells. CONCLUSION: Increased expression of G-CSF by injured arteries plays an essential role in contribution to recruitment and homing of MSCs to the site of the arterial lesion.


Assuntos
Fator Estimulador de Colônias de Granulócitos/metabolismo , Células-Tronco Mesenquimais/citologia , Lesões do Sistema Vascular/cirurgia , Lesões do Sistema Vascular/terapia , Angioplastia com Balão , Animais , Western Blotting , Artérias Carótidas/cirurgia , Diferenciação Celular , Movimento Celular , Células Cultivadas , Ensaio de Imunoadsorção Enzimática , Feminino , Imuno-Histoquímica , Hibridização in Situ Fluorescente , Masculino , Miócitos de Músculo Liso/citologia , Neointima/cirurgia , Neointima/terapia , Ratos , Ratos Sprague-Dawley , Reação em Cadeia da Polimerase Via Transcriptase Reversa
19.
Int J Cardiol ; 145(3): 468-75, 2010 Dec 03.
Artigo em Inglês | MEDLINE | ID: mdl-20053468

RESUMO

BACKGROUND: Extracellular matrix (ECM) accumulation significantly contributes to in-stent restenosis. In this regard, transforming growth factor (TGF)-ß, a positive regulator of ECM deposition, may be implicated in in-stent restenosis. The goal of this study was to assess the effect of blockade of TGF-ß on stent-induced restenosis in porcine coronary arteries. METHODS: An adenovirus expressing the ectodomain of the TGF-ß type II receptor (AdTß-ExR) was applied onto a coronary arterial segment of a pig (n=10) using an Infiltrator, followed by stent deployment. Controls consisted of adenoviruses expressing ß-galactosidase (AdLacZ) or phosphate-buffered saline (PBS) applied onto the other segment (n=10) of the same pig. RESULTS: Computer-based pathological morphometric analysis of stented coronary arteries, performed 4 weeks after stenting, demonstrated no significant difference in morphometric parameters such as in-stent neointimal area and % area stenosis between the AdTß-ExR group and control (n=7 for each). However the AdTß-ExR group had increased neointimal cell density, infiltration of inflammatory cells mostly consisting of CD3+ T cell, accumulation of hyaluronan, cell proliferation rate, and adventitial matrix metalloproteinase-1 (MMP-1) expression compared with control. The expression of connective tissue growth factor mRNA, measured by reverse transcription PCR, in cultured rat arterial smooth muscle cells was inhibited by AdTß-ExR at moi 60. CONCLUSIONS: Blockade of TGF-ß by catheter-based local intravascular gene delivery does not reduce stent-induced neointima formation 4 weeks after stenting in spite of modest inhibition of ECM accumulation, but it induces vascular inflammation and associated pathological changes that may potentially aggravate lesion progression.


Assuntos
Doença da Artéria Coronariana/terapia , Técnicas de Transferência de Genes , Neointima/terapia , Proteínas Serina-Treonina Quinases/genética , Receptores de Fatores de Crescimento Transformadores beta/genética , Stents/efeitos adversos , Fator de Crescimento Transformador beta/antagonistas & inibidores , Angioplastia , Animais , Complexo CD3/metabolismo , Cateterismo , Células Cultivadas , Fator de Crescimento do Tecido Conjuntivo/genética , Feminino , Ácido Hialurônico/metabolismo , Técnicas In Vitro , Masculino , Metaloproteinase 1 da Matriz/metabolismo , Músculo Liso Vascular/citologia , Músculo Liso Vascular/fisiologia , Ratos , Ratos Sprague-Dawley , Receptor do Fator de Crescimento Transformador beta Tipo II , Suínos , Linfócitos T/citologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA