Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 132
Filtrar
1.
J Cardiovasc Transl Res ; 13(2): 181-190, 2020 04.
Artigo em Inglês | MEDLINE | ID: mdl-31712976

RESUMO

The long-term patency rate of saphenous vein (SV) grafts is poor compared to arterial grafts. To investigate the effects of surgical preparation (distention) of SV on hydrogen sulfide (H2S) released from the endothelium, human SV segments were harvested from 43 patients during coronary artery bypass surgery (CABG). Acetylcholine (ACh) induced relaxation that was inhibited by NG-nitro-L-arginine + indomethacin and cysteine aminotransferase inhibitor aminooxyacetic acid in the normal SV. In contrast, ACh did not evoke relaxation in the distended SV (DSV). The concentration of H2S quantified by methylene blue assay in DSV was significantly lower than that in control. Transmission electron microscope and immunohistochemistry studies showed that the preparation destroyed the endothelium, smooth muscle, organelle, and vasa vasorum. We conclude that surgical preparation injures the endothelium and smooth muscle of the SV grafts and reduces H2S release from SV. These effects may contribute to the poor long-term patency of the SV graft.


Assuntos
Ponte de Artéria Coronária/efeitos adversos , Endotélio Vascular/transplante , Oclusão de Enxerto Vascular/etiologia , Sulfeto de Hidrogênio/metabolismo , Músculo Liso Vascular/transplante , Veia Safena/transplante , Coleta de Tecidos e Órgãos/efeitos adversos , Lesões do Sistema Vascular/etiologia , Idoso , Endotélio Vascular/lesões , Endotélio Vascular/metabolismo , Endotélio Vascular/fisiopatologia , Feminino , Oclusão de Enxerto Vascular/metabolismo , Oclusão de Enxerto Vascular/fisiopatologia , Humanos , Masculino , Pessoa de Meia-Idade , Músculo Liso Vascular/lesões , Músculo Liso Vascular/metabolismo , Músculo Liso Vascular/fisiopatologia , Veia Safena/lesões , Veia Safena/metabolismo , Veia Safena/fisiopatologia , Transdução de Sinais , Grau de Desobstrução Vascular , Lesões do Sistema Vascular/metabolismo , Lesões do Sistema Vascular/fisiopatologia
2.
J Vasc Surg ; 68(6S): 201S-207S, 2018 12.
Artigo em Inglês | MEDLINE | ID: mdl-29804740

RESUMO

OBJECTIVE: The main objective of this study was to define a role of sphingosine-1-phosphate receptor 1 (S1PR1) in the arterial injury response of a human artery. The hypotheses were tested that injury induces an expansion of S1PR1-positive cells and that these cells accumulate toward the lumen because they follow the sphingosine-1-phosphate gradient from arterial wall tissue (low) to plasma (high). METHODS: A humanized rat model was used in which denuded human internal mammary artery (IMA) was implanted into the position of the abdominal aorta of immunosuppressed Rowett nude rats. This injury model is characterized by medial as well as intimal hyperplasia, whereby intimal cells are of human origin. At 7, 14, and 28 days after implantation, grafts were harvested and processed for fluorescent immunostaining for S1PR1 and smooth muscle α-actin. Nuclei were stained with 4',6-diamidine-2'-phenylindole dihydrochloride. Using digitally reconstructed, complete cross sections of grafts, intimal and medial areas were measured, whereby the medial area had virtually been divided into an outer (toward adventitia) and inner (toward lumen) layer. The fraction of S1PR1-positive cells was determined in each layer by counting S1PR1-positive and S1PR1-negative cells. RESULTS: The fraction of S1PR1-postive cells in naive IMA is 58.9% ± 6.0% (mean ± standard deviation). At day 28 after implantation, 81.6% ± 4.4% of medial cells were scored S1PR1 positive (P < .01). At day 14, the ratio between S1PR1-positive and S1PR1-negative cells was significantly higher in the lumen-oriented inner layer (9.3 ± 2.1 vs 6.0 ± 1.0; P < .01). Cells appearing in the intima at day 7 and day 14 were almost all S1PR1 positive. At day 28, however, about one-third of intimal cells were scored S1PR1 negative. CONCLUSIONS: From these data, we conclude that denudation of IMA specifically induces the expansion of S1PR1-positive cells. Based on the nonrandom distribution of S1PR1-positive cells, we consider the possibility that much like lymphocytes, S1PR1-positive smooth muscle cells also use S1PR1 to recognize the sphingosine-1-phosphate gradient from tissue (low) to plasma (high) and so migrate out of the media toward the intima of the injured IMA.


Assuntos
Aorta Abdominal/cirurgia , Oclusão de Enxerto Vascular/metabolismo , Artéria Torácica Interna/transplante , Músculo Liso Vascular/transplante , Miócitos de Músculo Liso/transplante , Neointima , Receptores de Lisoesfingolipídeo/metabolismo , Animais , Movimento Celular , Proliferação de Células , Modelos Animais de Doenças , Oclusão de Enxerto Vascular/etiologia , Oclusão de Enxerto Vascular/patologia , Humanos , Lisofosfolipídeos/metabolismo , Masculino , Artéria Torácica Interna/metabolismo , Artéria Torácica Interna/patologia , Músculo Liso Vascular/metabolismo , Músculo Liso Vascular/patologia , Miócitos de Músculo Liso/metabolismo , Miócitos de Músculo Liso/patologia , Ratos Nus , Transdução de Sinais , Esfingosina/análogos & derivados , Esfingosina/metabolismo , Receptores de Esfingosina-1-Fosfato , Fatores de Tempo
3.
Cardiovasc Res ; 114(4): 601-610, 2018 03 15.
Artigo em Inglês | MEDLINE | ID: mdl-29373656

RESUMO

Atherosclerosis is the underlying pathology of many cardiovascular diseases. The formation and rupture of atherosclerotic plaques in the coronary arteries results in angina and myocardial infarction. Venous coronary artery bypass grafts are designed to reduce the consequences of atherosclerosis in the coronary arteries by diverting blood flow around the atherosclerotic plaques. However, vein grafts suffer a high failure rate due to intimal thickening that occurs as a result of vascular cell injury and activation and can act as 'a soil' for subsequent atherosclerotic plaque formation. A clinically-proven method for the reduction of vein graft intimal thickening and subsequent major adverse clinical events is currently not available. Consequently, a greater understanding of the underlying mechanisms of intimal thickening may be beneficial for the design of future therapies for vein graft failure. Vein grafting induces inflammation and endothelial cell damage and dysfunction, that promotes vascular smooth muscle cell (VSMC) migration, and proliferation. Injury to the wall of the vein as a result of grafting leads to the production of chemoattractants, remodelling of the extracellular matrix and cell-cell contacts; which all contribute to the induction of VSMC migration and proliferation. This review focuses on the role of altered behaviour of VSMCs in the vein graft and some of the factors which critically lead to intimal thickening that pre-disposes the vein graft to further atherosclerosis and re-occurrence of symptoms in the patient.


Assuntos
Ponte de Artéria Coronária/efeitos adversos , Oclusão de Enxerto Vascular/patologia , Músculo Liso Vascular/patologia , Músculo Liso Vascular/transplante , Miócitos de Músculo Liso/patologia , Miócitos de Músculo Liso/transplante , Veia Safena/patologia , Veia Safena/transplante , Animais , Movimento Celular , Proliferação de Células , Oclusão de Enxerto Vascular/etiologia , Oclusão de Enxerto Vascular/metabolismo , Oclusão de Enxerto Vascular/fisiopatologia , Humanos , Músculo Liso Vascular/metabolismo , Miócitos de Músculo Liso/metabolismo , Neointima , Fenótipo , Fatores de Risco , Veia Safena/metabolismo , Transdução de Sinais , Falha de Tratamento , Remodelação Vascular
4.
Arterioscler Thromb Vasc Biol ; 37(11): 2026-2037, 2017 11.
Artigo em Inglês | MEDLINE | ID: mdl-28860223

RESUMO

Vascular smooth muscle cells (VSMCs) play a role in the development of vascular disease, for example, neointimal formation, arterial aneurysm, and Marfan syndrome caused by genetic mutations in VSMCs, but little is known about the mechanisms of the disease process. Advances in induced pluripotent stem cell technology have now made it possible to derive VSMCs from several different somatic cells using a selection of protocols. As such, researchers have set out to delineate key signaling processes involved in triggering VSMC gene expression to grasp the extent of gene regulatory networks involved in phenotype commitment. This technology has also paved the way for investigations into diseases affecting VSMC behavior and function, which may be treatable once an identifiable culprit molecule or gene has been repaired. Moreover, induced pluripotent stem cell-derived VSMCs are also being considered for their use in tissue-engineered blood vessels as they may prove more beneficial than using autologous vessels. Finally, while several issues remains to be clarified before induced pluripotent stem cell-derived VSMCs can become used in regenerative medicine, they do offer both clinicians and researchers hope for both treating and understanding vascular disease. In this review, we aim to update the recent progress on VSMC generation from stem cells and the underlying molecular mechanisms of VSMC differentiation. We will also explore how the use of induced pluripotent stem cell-derived VSMCs has changed the game for regenerative medicine by offering new therapeutic avenues to clinicians, as well as providing researchers with a new platform for modeling of vascular disease.


Assuntos
Diferenciação Celular , Células-Tronco Pluripotentes Induzidas/fisiologia , Desenvolvimento Muscular , Músculo Liso Vascular/fisiologia , Miócitos de Músculo Liso/fisiologia , Aneurisma/genética , Aneurisma/metabolismo , Aneurisma/patologia , Aneurisma/cirurgia , Animais , Linhagem Celular , Linhagem da Célula , Modelos Animais de Doenças , Genótipo , Humanos , Células-Tronco Pluripotentes Induzidas/metabolismo , Células-Tronco Pluripotentes Induzidas/transplante , Síndrome de Marfan/genética , Síndrome de Marfan/metabolismo , Síndrome de Marfan/patologia , Síndrome de Marfan/cirurgia , Músculo Liso Vascular/metabolismo , Músculo Liso Vascular/transplante , Miócitos de Músculo Liso/metabolismo , Miócitos de Músculo Liso/transplante , Neointima , Fenótipo , Medicina Regenerativa , Transplante de Células-Tronco , Engenharia Tecidual
5.
Acta Biomater ; 59: 58-67, 2017 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-28642016

RESUMO

The strategy of vascular tissue engineering is to create a vascular substitute by combining autologous vascular cells with a tubular-shaped biodegradable scaffold. We have previously developed a novel electrospun bilayered vascular scaffold that provides proper biological and biomechanical properties as well as structural configuration. In this study, we investigated the clinical feasibility of a cellularized vascular scaffold in a preclinical large animal model. We fabricated the cellularized vascular construct with autologous endothelial progenitor cell (EPC)-derived endothelial cells (ECs) and smooth muscle cells (SMCs) followed by a pulsatile bioreactor preconditioning. This fully cellularized vascular construct was tested in a sheep carotid arterial interposition model. After preconditioning, confluent and mature EC and SMC layers in the scaffold were achieved. The cellularized constructs sustained the structural integrity with a high degree of graft patency without eliciting an inflammatory response over the course of the 6-month period in sheep. Moreover, the matured EC coverage on the lumen and a thick smooth muscle layer were formed at 6months after transplantation. We demonstrated that electrospun bilayered vascular scaffolds in conjunction with autologous vascular cells may be a clinically applicable alternative to traditional prosthetic vascular graft substitutes. STATEMENT OF SIGNIFICANCE: This study demonstrates the utility of tissue engineering to provide platform technologies for rehabilitation of patients recovering from severe, devastating cardiovascular diseases. The long-term goal is to provide alternatives to vascular grafting using bioengineered blood vessels derived from an autologous cell source with a functionalized vascular scaffold. This novel bilayered vascular construct for engineering blood vessels is designed to offer "off-the-shelf" availability for clinical translation.


Assuntos
Bioprótese , Prótese Vascular , Células Progenitoras Endoteliais , Músculo Liso Vascular , Miócitos de Músculo Liso , Alicerces Teciduais/química , Animais , Implante de Prótese Vascular , Células Progenitoras Endoteliais/citologia , Células Progenitoras Endoteliais/metabolismo , Células Progenitoras Endoteliais/transplante , Músculo Liso Vascular/citologia , Músculo Liso Vascular/metabolismo , Músculo Liso Vascular/transplante , Miócitos de Músculo Liso/citologia , Miócitos de Músculo Liso/metabolismo , Miócitos de Músculo Liso/transplante , Ovinos
6.
Biomed Res Int ; 2017: 7473437, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28503573

RESUMO

Objectives. The aim of this study was to test the effects of collagen external scaffold (CES) in intimal hyperplasia of vein grafts and explore its underlying mechanisms. Methods. Thirty-six New Zealand white rabbits were randomized into no-graft group, graft group, and CES group. The rabbit arteriovenous graft model was established. In CES group, the vein graft was wrapped around with CES. The hemodynamic parameters of vein grafts were measured intraoperatively and 4 weeks after operation by ultrasonic examination. Histological characteristics of vein grafts were also evaluated 4 weeks later. The mRNA and protein levels of proliferating cell nuclear antigen (PCNA), active cleaved-caspase-3 (ClvCasp-3), and smooth muscle 22 alpha (SM22α) were measured 4 weeks later by quantitative real-time PCR and western blot. Results. CES significantly improved the hemodynamic stability of vein grafts, with higher blood velocity and blood flow. Similarly, CES also markedly mitigated intimal hyperplasia and inhibited dilatation of vein grafts. In CES group, the upexpression of PCNA and ClvCasp-3 and the downexpression of SM22α were inhibited. Conclusion. CES exerts beneficial effects in mitigating intimal hyperplasia and improving remodeling of autogenous vein grafts, which may be associated with reducing the proliferation and apoptosis and preserving the phenotype of VSMCs.


Assuntos
Colágeno/uso terapêutico , Hiperplasia/terapia , Transplantes/transplante , Veias/transplante , Animais , Apoptose/genética , Proliferação de Células/genética , Colágeno/química , Modelos Animais de Doenças , Humanos , Hiperplasia/patologia , Proteínas dos Microfilamentos/genética , Proteínas Musculares/genética , Músculo Liso Vascular/transplante , Antígeno Nuclear de Célula em Proliferação/genética , RNA Mensageiro/genética , Coelhos , Alicerces Teciduais
7.
Basic Res Cardiol ; 112(4): 41, 2017 07.
Artigo em Inglês | MEDLINE | ID: mdl-28540527

RESUMO

Ischemic heart disease is still the leading cause of death even with the advancement of pharmaceutical therapies and surgical procedures. Early vascularization in the ischemic heart is critical for a better outcome. Although stem cell therapy has great potential for cardiovascular regeneration, the ideal cell type and delivery method of cells have not been resolved. We tested a new approach of stem cell therapy by delivery of induced vascular progenitor cells (iVPCs) grown on polymer micro-bundle scaffolds in a rat model of myocardial infarction. iVPCs partially reprogrammed from vascular endothelial cells (ECs) had potent angiogenic potential and were able to simultaneously differentiate into vascular smooth muscle cells (SMCs) and ECs in 2D culture. Under hypoxic conditions, iVPCs also secreted angiogenic cytokines such as vascular endothelial growth factor (VEGF) and basic fibroblast growth factor (bFGF) as measured by enzyme-linked immunosorbent assay (ELISA). A longitudinal micro-scaffold made from poly(lactic-co-glycolic acid) was sufficient for the growth and delivery of iVPCs. Co-cultured ECs and SMCs aligned well on the micro-bundle scaffold similarly as in the vessels. 3D cell/polymer micro-bundles formed by iVPCs and micro-scaffolds were transplanted into the ischemic myocardium in a rat model of myocardial infarction (MI) with ligation of the left anterior descending artery. Our in vivo data showed that iVPCs on the micro-bundle scaffold had higher survival, and better retention and engraftment in the myocardium than free iVPCs. iVPCs on the micro-bundles promoted better cardiomyocyte survival than free iVPCs. Moreover, iVPCs and iVPC/polymer micro-bundles treatment improved cardiac function (ejection fraction and fractional shortening, endocardial systolic volume) measured by echocardiography, increased vessel density, and decreased infarction size [endocardial and epicardial infarct (scar) length] better than untreated controls at 8 weeks after MI. We conclude that iVPCs grown on a polymer micro-bundle scaffold are new promising approach for cell-based therapy designed for cardiovascular regeneration in ischemic heart disease.


Assuntos
Células Progenitoras Endoteliais/transplante , Ácido Láctico/química , Músculo Liso Vascular/transplante , Infarto do Miocárdio/cirurgia , Miocárdio/patologia , Miócitos de Músculo Liso/transplante , Neovascularização Fisiológica , Ácido Poliglicólico/química , Engenharia Tecidual/métodos , Alicerces Teciduais , Animais , Diferenciação Celular , Proliferação de Células , Sobrevivência Celular , Células Cultivadas , Técnicas de Cocultura , Modelos Animais de Doenças , Células Progenitoras Endoteliais/metabolismo , Fator 2 de Crescimento de Fibroblastos/metabolismo , Músculo Liso Vascular/metabolismo , Infarto do Miocárdio/metabolismo , Infarto do Miocárdio/patologia , Infarto do Miocárdio/fisiopatologia , Miocárdio/metabolismo , Miócitos de Músculo Liso/metabolismo , Comunicação Parácrina , Fenótipo , Copolímero de Ácido Poliláctico e Ácido Poliglicólico , Ratos Sprague-Dawley , Transdução de Sinais , Fatores de Tempo , Fator A de Crescimento do Endotélio Vascular/metabolismo , Remodelação Ventricular
8.
Cardiovasc Res ; 113(5): 488-497, 2017 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-28137944

RESUMO

AIMS: Mechanical factors play significant roles in neointimal hyperplasia after vein grafting, but the mechanisms are not fully understood. Here, we investigated the roles of microRNA-33 (miR-33) in neointimal hyperplasia induced by arterial mechanical stretch after vein grafting. METHODS AND RESULTS: Grafted veins were generated by the 'cuff' technique. Neointimal hyperplasia and cell proliferation was significantly increased, and miR-33 expression was decreased after 1-, 2-, and 4-week grafts. In contrast, the expression of bone morphogenetic protein 3 (BMP3), which is a putative target of miR-33, and the phosphorylation of smad2 and smad5, which are potential downstream targets of BMP3, were increased in the grafted veins. miR-33 mimics/inhibitor and dual luciferase reporter assay confirmed the interaction of miR-33 and BMP3. miR-33 mimics attenuated, while miR-33 inhibitor accelerated, proliferation of venous smooth muscle cells (SMCs). Moreover, recombinant BMP3 increased SMC proliferation and P-smad2 and P-smad5 levels, whereas BMP3-directed siRNAs had the opposite effect. Then, venous SMCs were exposed to a 10%-1.25 Hz cyclic stretch (arterial stretch) by using the FX4000 cyclic stretch loading system in vitro to mimic arterial mechanical conditions. The arterial stretch increased venous SMC proliferation and repressed miR-33 expression, but enhanced BMP3 expression and smad2 and smad5 phosphorylation. Furthermore, perivascular multi-point injection in vivo demonstrated that agomiR-33 not only attenuates BMP3 expression and smad2 and smad5 phosphorylation, but also slows neointimal formation and cell proliferation in grafted veins. These effects of agomiR-33 on grafted veins could be reversed by local injection of BMP3 lentivirus. CONCLUSION: The miR-33-BMP3-smad signalling pathway protects against venous SMC proliferation in response to the arterial stretch. miR-33 is a target that attenuates neointimal hyperplasia in grafted vessels and may have potential clinical applications.


Assuntos
Proliferação de Células , Veias Jugulares/metabolismo , Veias Jugulares/transplante , Mecanotransdução Celular , MicroRNAs/metabolismo , Neointima , Regiões 3' não Traduzidas , Animais , Antagomirs/genética , Antagomirs/metabolismo , Sítios de Ligação , Proteína Morfogenética Óssea 3/genética , Proteína Morfogenética Óssea 3/metabolismo , Células Cultivadas , Hiperplasia , Veias Jugulares/patologia , Masculino , MicroRNAs/genética , Músculo Liso Vascular/metabolismo , Músculo Liso Vascular/patologia , Músculo Liso Vascular/transplante , Miócitos de Músculo Liso/metabolismo , Miócitos de Músculo Liso/patologia , Miócitos de Músculo Liso/transplante , Fosforilação , Interferência de RNA , Ratos Sprague-Dawley , Proteína Smad2/metabolismo , Proteína Smad5/metabolismo , Estresse Mecânico , Fatores de Tempo , Transfecção
9.
J Vasc Surg ; 66(3): 883-890.e1, 2017 09.
Artigo em Inglês | MEDLINE | ID: mdl-28017585

RESUMO

OBJECTIVE: One of the rate-limiting barriers within the field of vascular tissue engineering is the lengthy fabrication time associated with expanding appropriate cell types in culture. One particularly attractive cell type for this purpose is the adipose-derived mesenchymal stem cell (AD-MSC), which is abundant and easily harvested from liposuction procedures. Even this cell type has its drawbacks, however, including the required culture period for expansion, which could pose risks of cellular transformation or contamination. Eliminating culture entirely would be ideal to avoid these concerns. In this study, we used the raw population of cells obtained after digestion of human liposuction aspirates, known as the stromal vascular fraction (SVF), as an abundant, culture-free cell source for tissue-engineered vascular grafts (TEVGs). METHODS: SVF cells and donor-paired cultured AD-MSCs were first assessed for their abilities to differentiate into vascular smooth muscle cells (SMCs) after angiotensin II stimulation and to secrete factors (eg, conditioned media) that promote SMC migration. Next, both cell types were incorporated into TEVG scaffolds, implanted as an aortic graft in a Lewis rat model, and assessed for their patency and composition. RESULTS: In general, the human SVF cells were able to perform the same functions as AD-MSCs isolated from the same donor by culture expansion. Specifically, cells within the SVF performed two important functions; namely, they were able to differentiate into SMCs (SVF calponin expression: 16.4% ± 7.7% vs AD-MSC: 19.9%% ± 1.7%) and could secrete promigratory factors (SVF migration rate relative to control: 3.1 ± 0.3 vs AD-MSC: 2.5 ± 0.5). The SVF cells were also capable of being seeded within biodegradable, elastomeric, porous scaffolds that, when implanted in vivo for 8 weeks, generated patent TEVGs (SVF: 83% patency vs AD-MSC: 100% patency) populated with primary vascular components (eg, SMCs, endothelial cells, collagen, and elastin). CONCLUSIONS: Human adipose tissue can be used as a culture-free cell source to create TEVGs, laying the groundwork for the rapid production of cell-seeded grafts.


Assuntos
Tecido Adiposo/irrigação sanguínea , Bioprótese , Implante de Prótese Vascular/instrumentação , Prótese Vascular , Músculo Liso Vascular/transplante , Miócitos de Músculo Liso/transplante , Células Estromais/transplante , Engenharia Tecidual/métodos , Adulto , Angiotensina II/farmacologia , Animais , Aorta Abdominal/metabolismo , Aorta Abdominal/patologia , Aorta Abdominal/cirurgia , Implante de Prótese Vascular/métodos , Diferenciação Celular , Movimento Celular , Separação Celular , Células Cultivadas , Feminino , Humanos , Lipectomia , Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais/efeitos dos fármacos , Células-Tronco Mesenquimais/metabolismo , Músculo Liso Vascular/efeitos dos fármacos , Músculo Liso Vascular/metabolismo , Miócitos de Músculo Liso/efeitos dos fármacos , Miócitos de Músculo Liso/metabolismo , Fenótipo , Ratos Endogâmicos Lew , Células Estromais/efeitos dos fármacos , Células Estromais/metabolismo , Fatores de Tempo , Alicerces Teciduais
10.
Arterioscler Thromb Vasc Biol ; 36(11): 2167-2175, 2016 11.
Artigo em Inglês | MEDLINE | ID: mdl-27659097

RESUMO

OBJECTIVE: Plasminogen activator inhibitor-1 (PAI-1), a serine protease inhibitor that promotes and inhibits cell migration, plays a complex and important role in adverse vascular remodeling. Little is known about the effects of pharmacological PAI-1 inhibitors, an emerging drug class, on migration of vascular smooth muscle cells (SMCs) and endothelial cells (ECs), crucial mediators of vascular remodeling. We investigated the effects of PAI-039 (tiplaxtinin), a specific PAI-1 inhibitor, on SMC and EC migration in vitro and vascular remodeling in vivo. APPROACH AND RESULTS: PAI-039 inhibited SMC migration through collagen gels, including those supplemented with vitronectin and other extracellular matrix proteins, but did not inhibit migration of PAI-1-deficient SMCs, suggesting that its antimigratory effects were PAI-1-specific and physiologically relevant. However, PAI-039 did not inhibit EC migration. PAI-039 inhibited phosphorylation and nuclear translocation of signal transducers and activators of transcription-1 in SMCs, but had no discernable effect on signal transducer and activator of transcription-1 signaling in ECs. Expression of low-density lipoprotein receptor-related protein 1, a motogenic PAI-1 receptor that activates Janus kinase/signal transducers and activators of transcription-1 signaling, was markedly lower in ECs than in SMCs. Notably, PAI-039 significantly inhibited intimal hyperplasia and inflammation in murine models of adverse vascular remodeling, but did not adversely affect re-endothelialization after endothelium-denuding mechanical vascular injury. CONCLUSIONS: PAI-039 inhibits SMC migration and intimal hyperplasia, while having no inhibitory effect on ECs, which seems to be because of differences in PAI-1-dependent low-density lipoprotein receptor-related protein 1/Janus kinase/signal transducer and activator of transcription-1 signaling between SMCs and ECs. These findings suggest that PAI-1 may be an important therapeutic target in obstructive vascular diseases characterized by neointimal hyperplasia.


Assuntos
Lesões das Artérias Carótidas/tratamento farmacológico , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Ácidos Indolacéticos/farmacologia , Músculo Liso Vascular/efeitos dos fármacos , Músculo Liso/efeitos dos fármacos , Neointima , Inibidor 1 de Ativador de Plasminogênio/metabolismo , Inibidores de Serina Proteinase/farmacologia , Animais , Lesões das Artérias Carótidas/genética , Lesões das Artérias Carótidas/metabolismo , Lesões das Artérias Carótidas/patologia , Células Cultivadas , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/metabolismo , Células Endoteliais/patologia , Genótipo , Humanos , Hiperplasia , Janus Quinases/metabolismo , Proteína-1 Relacionada a Receptor de Lipoproteína de Baixa Densidade , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Terapia de Alvo Molecular , Músculo Liso/metabolismo , Músculo Liso/patologia , Músculo Liso/transplante , Músculo Liso Vascular/metabolismo , Músculo Liso Vascular/patologia , Músculo Liso Vascular/transplante , Fenótipo , Fosforilação , Inibidor 1 de Ativador de Plasminogênio/deficiência , Inibidor 1 de Ativador de Plasminogênio/genética , Reepitelização/efeitos dos fármacos , Receptores de LDL/deficiência , Receptores de LDL/genética , Fator de Transcrição STAT1/metabolismo , Transdução de Sinais/efeitos dos fármacos , Proteínas Supressoras de Tumor/deficiência , Proteínas Supressoras de Tumor/genética , Remodelação Vascular/efeitos dos fármacos , Veia Cava Inferior/efeitos dos fármacos , Veia Cava Inferior/metabolismo , Veia Cava Inferior/patologia , Veia Cava Inferior/transplante
11.
Arterioscler Thromb Vasc Biol ; 36(6): 1230-9, 2016 06.
Artigo em Inglês | MEDLINE | ID: mdl-27079875

RESUMO

OBJECTIVE: Smooth muscle-like cells are major cell components of transplant arteriosclerosis lesions. This study investigated the origin of the smooth muscle-like cells, the mechanisms responsible for their accumulation in the neointima, and the factors that drive these processes. APPROACH AND RESULTS: A murine aortic transplantation model was established by transplanting miR-155(-/-) bone marrow cells into miR-155(+/+) mice. MicroRNA-155 was found to play a functional role in the transplant arteriosclerosis. Moreover, we found that the nonbone marrow-derived progenitor cells with markers of both early differentiated smooth muscles and stem cells in the allograft adventitia were smooth muscle progenitor cells. Purified smooth muscle progenitor cells expressed a mature smooth muscle cell marker when induced by platelet-derived growth factor-BB in vitro. In vivo, these cells could migrate into the intima from the adventitia and could contribute to the neointimal hyperplasia. The loss of microRNA-155 in bone marrow-derived cells decreased the concentration gradient of monocyte chemoattractant protein 1 between the intima and the adventitia of the allografts, which reduced the migration of smooth muscle progenitor cells from the adventitia into the neointima. CONCLUSIONS: This study demonstrated that microRNA-155 promoted the directional migration of smooth muscle progenitor cells from the adventitia by regulating the monocyte chemoattractant protein 1 concentration gradient, which aggravated transplant arteriosclerosis.


Assuntos
Aorta/transplante , Aterosclerose/metabolismo , Quimiocina CCL2/metabolismo , Quimiotaxia , MicroRNAs/metabolismo , Músculo Liso Vascular/transplante , Miócitos de Músculo Liso/transplante , Células-Tronco/metabolismo , Enxerto Vascular/efeitos adversos , Túnica Adventícia/metabolismo , Túnica Adventícia/patologia , Aloenxertos , Animais , Aorta/metabolismo , Aorta/patologia , Aterosclerose/etiologia , Aterosclerose/genética , Aterosclerose/patologia , Transplante de Medula Óssea , Diferenciação Celular , Células Cultivadas , Genótipo , Hiperplasia , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , MicroRNAs/genética , Músculo Liso Vascular/metabolismo , Músculo Liso Vascular/patologia , Miócitos de Músculo Liso/metabolismo , Miócitos de Músculo Liso/patologia , Neointima , Fenótipo , Transdução de Sinais , Células-Tronco/patologia , Fatores de Tempo , Transfecção
12.
Transplantation ; 100(8): 1656-66, 2016 08.
Artigo em Inglês | MEDLINE | ID: mdl-27014792

RESUMO

BACKGROUND: Development of transplant vasculopathy is a major cause of graft loss and mortality in solid organ transplant recipients. Previous studies in mice have indicated that vanin-1, a member of the vanin protein family with pantetheinase activity, is possibly involved in neointima formation. Here, we investigated if RR6, a recently developed vanin inhibitor, could attenuate development of transplant vasculopathy. METHODS: Abdominal allogeneic aorta transplantation from Dark Agouti to Brown Norway rats was performed. Surface neointima was quantified 2 and 4 weeks after transplantation. Systemic vanin activity was measured, and allograft leukocyte infiltration, glutathione-synthesizing capacity, matrix metalloproteinase 9 expression and neointimal smooth muscle cell (SMC) proliferation were assessed by immunohistochemistry. In vitro, the effects of RR6 on SMC proliferation (water-soluble tetrazolium-1 assay) and cytokine-induced apoptosis (flow cytometry) were investigated. RESULTS: RR6 treatment significantly reduced systemic pantetheinase activity during the 4-week follow-up period. RR6 attenuated neointima formation 4 weeks after transplantation. Neointimal SMC proliferation and medial SMC matrix metalloproteinase 9 expression were not altered by RR6. However, RR6 significantly reduced neointimal macrophage influx that was accompanied by increased GCLC messenger RNA expression. In vitro, RR6 inhibited platelet-derived growth factor-induced SMC proliferation and protected SMCs from TNF-α-induced apoptosis. CONCLUSIONS: Pharmacological inhibition of vanin activity attenuates development of transplant vasculopathy. This was accompanied by reduced macrophage infiltration and increased glutathione-synthesizing capacity. In vitro, RR6 reduced SMC proliferation and apoptosis that was not confirmed in vivo. Further in-depth studies are warranted to reveal the underlying mechanism(s) of RR6-induced attenuation of transplant vasculopathy in vivo.


Assuntos
Amidoidrolases/antagonistas & inibidores , Aorta Abdominal/efeitos dos fármacos , Aorta Abdominal/transplante , Doenças da Aorta/prevenção & controle , Inibidores Enzimáticos/farmacologia , Sobrevivência de Enxerto/efeitos dos fármacos , Amidoidrolases/metabolismo , Animais , Aorta Abdominal/enzimologia , Aorta Abdominal/patologia , Doenças da Aorta/enzimologia , Doenças da Aorta/patologia , Apoptose/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Modelos Animais de Doenças , Proteínas Ligadas por GPI/antagonistas & inibidores , Proteínas Ligadas por GPI/metabolismo , Glutationa/metabolismo , Humanos , Macrófagos/efeitos dos fármacos , Macrófagos/metabolismo , Masculino , Músculo Liso Vascular/efeitos dos fármacos , Músculo Liso Vascular/enzimologia , Músculo Liso Vascular/patologia , Músculo Liso Vascular/transplante , Miócitos de Músculo Liso/efeitos dos fármacos , Miócitos de Músculo Liso/enzimologia , Miócitos de Músculo Liso/patologia , Neointima , Ratos Endogâmicos BN , Transdução de Sinais/efeitos dos fármacos , Fatores de Tempo , Transplante Homólogo
13.
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
14.
Exp Clin Transplant ; 14(1): 86-92, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26862826

RESUMO

OBJECTIVES: Methods for conservation and preservation of vascular grafts are often controversially discussed. Furthermore, immunologic monitoring or immunotherapy for allogeneic graft is not considered necessary in many cases. The present study was initiated to examine the cellular vitality and functional efficiency of vein transplant during preservation. MATERIALS AND METHODS: Twenty-seven human vein segments (vena saphena magna) were stored after explant in University of Wisconsin solution or histidine-tryptophan-ketoglutarate solution at 4 °C. After 3, 24, 48, 72, and 96 hours, vein functionality was tested. Ring segments were fixed by triangles in Krebs-Henseleit buffer. Contractile function was measured after addition of potassium chloride solution (80 mM) and phenylephrine (0.2, 2, or 20 µM). To investigate endothelium-dependent vasorelaxation, 1 µM acetylcholine was added. RESULTS: Of 27 segments, 5 showed endothelium-dependent relaxation. Vasorelaxation continued for up to 48 hours after administration of acetylcholine in University of Wisconsin solution and for up to 24 hours in histidine-tryptophane-ketoglutarate solution. At 48 hours, potassium chloride solution-induced vasocontraction was 17% more effective than phenylephrine in University of Wisconsin solution. University of Wisconsin solution was significantly more effective than histidine-tryptophane-ketoglutarate solution in terms of preservation of phenylephrine (0.2, 2 µM)-induced vasocontraction. Phenylephrine (2 µM)-induced contraction was retained in University of Wisconsin solution after 24 hours by 81% and after 48 hours by 55%, with comparable results in histidine-tryptophane-ketoglutarate solution of only 62% and 34% after 24 and 48 hours. CONCLUSIONS: At 48 hours, human saphenous vein transplants had better endothelium and smooth muscle function when preserved in University of Wisconsin solution versus histidine-tryptophane-ketoglutarate solution.


Assuntos
Endotélio Vascular/efeitos dos fármacos , Músculo Liso Vascular/efeitos dos fármacos , Soluções para Preservação de Órgãos/farmacologia , Veia Safena/efeitos dos fármacos , Preservação de Tecido/métodos , Adenosina/farmacologia , Alopurinol/farmacologia , Temperatura Baixa , Relação Dose-Resposta a Droga , Endotélio Vascular/transplante , Endotélio Vascular/ultraestrutura , Glucose/farmacologia , Glutationa/farmacologia , Humanos , Insulina/farmacologia , Manitol/farmacologia , Músculo Liso Vascular/transplante , Músculo Liso Vascular/ultraestrutura , Cloreto de Potássio/farmacologia , Procaína/farmacologia , Rafinose/farmacologia , Veia Safena/transplante , Veia Safena/ultraestrutura , Fatores de Tempo , Coleta de Tecidos e Órgãos , Vasoconstrição/efeitos dos fármacos , Vasoconstritores/farmacologia , Vasodilatação/efeitos dos fármacos , Vasodilatadores/farmacologia
15.
J Vasc Surg ; 64(5): 1444-1449, 2016 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26432280

RESUMO

BACKGROUND: The production of growth factors from several experimental arterial conduits was determined. METHODS: We implanted 105 experimental arterial grafts that were 1 cm long in the abdominal aorta of Lewis rats (average weight, 250 g). Five different types of grafts were analyzed: arterial isografts, vein grafts, arterial allografts, and polytetrafluoroethylene (PTFE) grafts with normal or decreased compliance. Animals were killed humanely 4 weeks after surgery and the production of platelet-derived growth factor (PDGF), basic fibroblast growth factor (bFGF), transforming growth factor-ß, tumor necrosis factor-α, and interleukin-1 was analyzed. RESULTS: Myointimal hyperplasia (MH) was evident in vein grafts, arterial allografts, and PTFE grafts, but not in arterial isografts. Growth factor production was increased for grafts prone to develop MH like vein, PTFE grafts, and arterial allografts. PDGF and bFGF were increased significantly for PTFE and vein grafts, but not for arterial allografts. The importance of bFGF and PGDF was confirmed by the capability of antibody to PDGF and to bFGF to reduce the mitogenic activity of smooth muscle cells, in vivo and in vitro, for PTFE and vein grafts, but not for arterial allografts, in which a predominant role was played by interleukin-1 and tumor necrosis factor-α. CONCLUSIONS: Agents able to neutralize this increased production of growth factors, either directly or by competition with their receptors, can prevent MH formation.


Assuntos
Aorta Abdominal/cirurgia , Artérias/transplante , Implante de Prótese Vascular/instrumentação , Prótese Vascular , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Veias/transplante , Aloenxertos , Animais , Aorta Abdominal/metabolismo , Aorta Abdominal/patologia , Artérias/metabolismo , Artérias/patologia , Proliferação de Células , Células Cultivadas , Meios de Cultivo Condicionados/metabolismo , Fator 2 de Crescimento de Fibroblastos/metabolismo , Hiperplasia , Interleucina-1/metabolismo , Isoenxertos , Modelos Animais , Músculo Liso Vascular/metabolismo , Músculo Liso Vascular/patologia , Músculo Liso Vascular/transplante , Miócitos de Músculo Liso/metabolismo , Miócitos de Músculo Liso/patologia , Miócitos de Músculo Liso/transplante , Neointima , Fator de Crescimento Derivado de Plaquetas/metabolismo , Politetrafluoretileno , Desenho de Prótese , Ratos Endogâmicos Lew , Fator de Crescimento Transformador beta/metabolismo , Fator de Necrose Tumoral alfa/metabolismo , Veias/metabolismo , Veias/patologia
16.
Transplantation ; 100(1): 80-90, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26674731

RESUMO

BACKGROUND: Transplant vasculopathy (TV) is a major cause for late graft loss after cardiac transplantation. Endothelial damage and T cell infiltration play a pivotal role in the development of TV. Because N-octanoyl dopamine (NOD) inhibits vascular inflammation and suppresses T cell activation in vitro, we here tested the hypothesis that NOD treatment ameliorates TV. METHODS: Aortic grafts were orthotopically transplanted in the Dark Agouti to Brown Norway strain combination. Recipient rats were treated with NOD or vehicle administered via osmotic minipumps. Histology and quantitative polymerase chain reaction (qPCR) were performed on nontransplanted aortas and grafts explanted 2 and 4 weeks after transplantation to assess the degree of TV, inflammation, apoptosis, and number of (proliferating) α smooth muscle actin (αSMA) neointimal cells. In vitro analyses of human aortic smooth muscle cells were performed to test the effect of NOD on proliferation (WST-1 assay), cell cycle (flow cytometry and qPCR), and cytokine-induced apoptosis (flow cytometry). RESULTS: Allografts from vehicle-treated recipients developed neointimal lesions predominantly consisting of αSMA-expressing cells. NOD treatment significantly reduced neointima formation and neointimal αSMA cells. In situ, smooth muscle cell proliferation (Ki67) was not influenced by NOD. Macrophage (CD68), T (CD3), and Natural Killer (ANK61) cell infiltration as well as intragraft TNFα and IFNγ mRNA expression were similar in both groups. Medial apoptosis (cleaved caspase-3) was significantly reduced by NOD. In vitro, NOD inhibited proliferation of human aortic smooth muscle cells by causing a G1-arrest and protected from TNFα-induced apoptosis. CONCLUSIONS: This study identified NOD as potential treatment modality to attenuate TV. Our data clearly support a vasculoprotective effect of NOD by reducing smooth muscle cell proliferation and inflammation-induced apoptosis.


Assuntos
Anti-Inflamatórios/farmacologia , Doenças da Aorta/prevenção & controle , Dopamina/análogos & derivados , Músculo Liso Vascular/efeitos dos fármacos , Músculo Liso Vascular/transplante , Miócitos de Músculo Liso/efeitos dos fármacos , Miócitos de Músculo Liso/transplante , Actinas/metabolismo , Aloenxertos , Animais , Anti-Inflamatórios/administração & dosagem , Aorta/efeitos dos fármacos , Aorta/metabolismo , Aorta/patologia , Aorta/transplante , Doenças da Aorta/metabolismo , Doenças da Aorta/patologia , Apoptose/efeitos dos fármacos , Ciclo Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Quimiotaxia de Leucócito/efeitos dos fármacos , Citoproteção , Dopamina/administração & dosagem , Dopamina/farmacologia , Humanos , Mediadores da Inflamação/metabolismo , Bombas de Infusão Implantáveis , Células Matadoras Naturais/efeitos dos fármacos , Células Matadoras Naturais/imunologia , Células Matadoras Naturais/metabolismo , Macrófagos/efeitos dos fármacos , Macrófagos/imunologia , Macrófagos/metabolismo , Masculino , Músculo Liso Vascular/metabolismo , Músculo Liso Vascular/patologia , Miócitos de Músculo Liso/metabolismo , Miócitos de Músculo Liso/patologia , Neointima , Ratos Endogâmicos BN , Linfócitos T/efeitos dos fármacos , Linfócitos T/imunologia , Linfócitos T/metabolismo , Fatores de Tempo
17.
Nat Cell Biol ; 17(8): 994-1003, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-26214132

RESUMO

The use of human pluripotent stem cells for in vitro disease modelling and clinical applications requires protocols that convert these cells into relevant adult cell types. Here, we report the rapid and efficient differentiation of human pluripotent stem cells into vascular endothelial and smooth muscle cells. We found that GSK3 inhibition and BMP4 treatment rapidly committed pluripotent cells to a mesodermal fate and subsequent exposure to VEGF-A or PDGF-BB resulted in the differentiation of either endothelial or vascular smooth muscle cells, respectively. Both protocols produced mature cells with efficiencies exceeding 80% within six days. On purification to 99% via surface markers, endothelial cells maintained their identity, as assessed by marker gene expression, and showed relevant in vitro and in vivo functionality. Global transcriptional and metabolomic analyses confirmed that the cells closely resembled their in vivo counterparts. Our results suggest that these cells could be used to faithfully model human disease.


Assuntos
Diferenciação Celular , Linhagem da Célula , Células Endoteliais/fisiologia , Células-Tronco Pluripotentes Induzidas/fisiologia , Músculo Liso Vascular/fisiologia , Miócitos de Músculo Liso/fisiologia , Animais , Becaplermina , Biomarcadores/metabolismo , Proteína Morfogenética Óssea 4/farmacologia , Diferenciação Celular/efeitos dos fármacos , Linhagem Celular , Linhagem da Célula/efeitos dos fármacos , Técnicas de Cocultura , Relação Dose-Resposta a Droga , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/enzimologia , Células Endoteliais/transplante , Perfilação da Expressão Gênica/métodos , Regulação da Expressão Gênica no Desenvolvimento , Quinase 3 da Glicogênio Sintase/antagonistas & inibidores , Quinase 3 da Glicogênio Sintase/metabolismo , Glicogênio Sintase Quinase 3 beta , Células Endoteliais da Veia Umbilical Humana/fisiologia , Humanos , Células-Tronco Pluripotentes Induzidas/efeitos dos fármacos , Células-Tronco Pluripotentes Induzidas/enzimologia , Células-Tronco Pluripotentes Induzidas/transplante , Metabolômica/métodos , Camundongos Endogâmicos NOD , Camundongos SCID , Músculo Liso Vascular/citologia , Músculo Liso Vascular/efeitos dos fármacos , Músculo Liso Vascular/enzimologia , Músculo Liso Vascular/transplante , Miócitos de Músculo Liso/efeitos dos fármacos , Miócitos de Músculo Liso/enzimologia , Miócitos de Músculo Liso/transplante , Neovascularização Fisiológica , Fenótipo , Inibidores de Proteínas Quinases/farmacologia , Proteínas Proto-Oncogênicas c-sis/farmacologia , Fatores de Tempo , Transcrição Gênica , Transfecção , Fator A de Crescimento do Endotélio Vascular/farmacologia , Via de Sinalização Wnt/efeitos dos fármacos
18.
Circ Res ; 116(8): 1392-412, 2015 Apr 10.
Artigo em Inglês | MEDLINE | ID: mdl-25858065

RESUMO

The vasculature plays an indispensible role in organ development and maintenance of tissue homeostasis, such that disturbances to it impact greatly on developmental and postnatal health. Although cell turnover in healthy blood vessels is low, it increases considerably under pathological conditions. The principle sources for this phenomenon have long been considered to be the recruitment of cells from the peripheral circulation and the re-entry of mature cells in the vessel wall back into cell cycle. However, recent discoveries have also uncovered the presence of a range of multipotent and lineage-restricted progenitor cells in the mural layers of postnatal blood vessels, possessing high proliferative capacity and potential to generate endothelial, smooth muscle, hematopoietic or mesenchymal cell progeny. In particular, the tunica adventitia has emerged as a progenitor-rich compartment with niche-like characteristics that support and regulate vascular wall progenitor cells. Preliminary data indicate the involvement of some of these vascular wall progenitor cells in vascular disease states, adding weight to the notion that the adventitia is integral to vascular wall pathogenesis, and raising potential implications for clinical therapies. This review discusses the current body of evidence for the existence of vascular wall progenitor cell subpopulations from development to adulthood and addresses the gains made and significant challenges that lie ahead in trying to accurately delineate their identities, origins, regulatory pathways, and relevance to normal vascular structure and function, as well as disease.


Assuntos
Doenças Cardiovasculares/patologia , Células Progenitoras Endoteliais/patologia , Músculo Liso Vascular/patologia , Mioblastos de Músculo Liso/patologia , Animais , Doenças Cardiovasculares/metabolismo , Doenças Cardiovasculares/cirurgia , Diferenciação Celular , Linhagem da Célula , Proliferação de Células , Células Progenitoras Endoteliais/metabolismo , Células Progenitoras Endoteliais/transplante , Humanos , Músculo Liso Vascular/metabolismo , Músculo Liso Vascular/transplante , Mioblastos de Músculo Liso/metabolismo , Mioblastos de Músculo Liso/transplante , Neovascularização Patológica , Neovascularização Fisiológica , Regeneração , Medicina Regenerativa/métodos , Nicho de Células-Tronco
19.
Interact Cardiovasc Thorac Surg ; 21(1): 50-4, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-25820759

RESUMO

OBJECTIVES: The excess proliferation of vascular smooth muscle cells (VSMCs) and the development of intimal hyperplasia is a hallmark of vein graft failure. This study aimed to verify that a single intraoperative transfection of early growth response gene-1 (Egr-1) decoy oligonucleotide (ODN) can suppress vein graft proliferation of VSMCs and intimal hyperplasia. METHODS: In a rabbit model, jugular veins were treated with Egr-1 decoy ODN, scrambled decoy ODN, Fugene6, or were left untreated, then grafted to the carotid artery. The vein graft samples were obtained 48 h, 1, 2 or 3 weeks after surgery. The thickness of the intima and intima/media ratio in the grafts was analysed by haematoxylin-eosin (HE) staining. The expression of the Egr-1 decoy ODN transfected in the vein was analysed using fluorescent microscopy. Egr-1 mRNA was measured using reverse transcription-polymerase chain reaction. The expression of Egr-1 protein was analysed by Western blot and immunohistochemistry. RESULTS: Transfection efficiency of the ODN was confirmed by 4', 6-diamidino-2-phenylindole staining. In the grafts treated with Egr-1 decoy ODN, our study achieved statistically significant inhibition of intimal hyperplasia by ∼58% at 3 weeks. Transfection of Egr-1 decoy ODNs decreased the protein expression of Egr-1 and Egr-1 mRNA. CONCLUSIONS: We confirmed that gene therapy using in vivo transfection of an Egr-1 decoy ODN significantly inhibits proliferation of VSMC and intimal hyperplasia of vein grafts in a rabbit model.


Assuntos
Proliferação de Células , Proteína 1 de Resposta de Crescimento Precoce/metabolismo , Terapia Genética/métodos , Oclusão de Enxerto Vascular/prevenção & controle , Músculo Liso Vascular/transplante , Miócitos de Músculo Liso/transplante , Neointima , Oligonucleotídeos/metabolismo , Veia Safena/transplante , Animais , Modelos Animais de Doenças , Regulação para Baixo , Proteína 1 de Resposta de Crescimento Precoce/genética , Oclusão de Enxerto Vascular/genética , Oclusão de Enxerto Vascular/metabolismo , Oclusão de Enxerto Vascular/patologia , Hiperplasia , Masculino , Músculo Liso Vascular/metabolismo , Músculo Liso Vascular/patologia , Miócitos de Músculo Liso/metabolismo , Miócitos de Músculo Liso/patologia , Oligonucleotídeos/genética , RNA Mensageiro/metabolismo , Coelhos , Veia Safena/metabolismo , Veia Safena/patologia , Transdução de Sinais , Fatores de Tempo , Transfecção
20.
Microvasc Res ; 100: 9-16, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-25660475

RESUMO

The excessive proliferation of vascular smooth muscle cells was key factor in the restenosis of vein graft. And the Notch signaling was demonstrated to regulate vSMC proliferation and differentiation. Soluble Jagged-1 (sJag1) can inhibit Notch signaling in vitro and in vivo; however, its capacity to suppress restenosis of vein graft remains unknown. Under the microscope, the left jugular vein of these rats was interposed into the left common carotid artery, followed without any treatment (control), or with Ad-Jag1 (treatment) or placebo (DMSO) post operation. We showed that Ad-Jag1 can attenuate restenosis of vein graft by inducing decreased proliferation and increased apoptosis in vivo. Notch1-Hey2 signaling is critical for the development of intima thickening by controlling vSMC-fate determination. By blocking Notch signaling, Ad-Jag1 can significantly inhibit intima thickening. These studies identify that Ad-Jag1 can restore the vSMC phenotype and inhibit the vSMC proliferation by suppression of Notch1 signaling, and thus open a new avenue for the treatment of restenosis in vein graft.


Assuntos
Proteínas de Ligação ao Cálcio/biossíntese , Terapia Genética/métodos , Oclusão de Enxerto Vascular/prevenção & controle , Peptídeos e Proteínas de Sinalização Intercelular/biossíntese , Veias Jugulares/transplante , Proteínas de Membrana/biossíntese , Músculo Liso Vascular/transplante , Receptor Notch1/metabolismo , Transdução de Sinais , Animais , Apoptose , Proteínas de Ligação ao Cálcio/genética , Artéria Carótida Primitiva/cirurgia , Diferenciação Celular , Proliferação de Células , Células Cultivadas , Modelos Animais de Doenças , Oclusão de Enxerto Vascular/genética , Oclusão de Enxerto Vascular/metabolismo , Oclusão de Enxerto Vascular/patologia , Peptídeos e Proteínas de Sinalização Intercelular/genética , Proteína Jagged-1 , Veias Jugulares/metabolismo , Veias Jugulares/patologia , Masculino , Proteínas de Membrana/genética , Músculo Liso Vascular/metabolismo , Músculo Liso Vascular/patologia , Miócitos de Músculo Liso/metabolismo , Miócitos de Músculo Liso/patologia , Neointima , Fenótipo , Ratos Wistar , Proteínas Serrate-Jagged , Fatores de Tempo , Transfecção , Enxerto Vascular
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...