Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 91
Filtrar
1.
Nat Commun ; 11(1): 4311, 2020 08 27.
Artigo em Inglês | MEDLINE | ID: mdl-32855420

RESUMO

Pulmonary disease increases the risk of developing abdominal aortic aneurysms (AAA). However, the mechanism underlying the pathological dialogue between the lungs and aorta is undefined. Here, we find that inflicting acute lung injury (ALI) to mice doubles their incidence of AAA and accelerates macrophage-driven proteolytic damage of the aortic wall. ALI-induced HMGB1 leaks and is captured by arterial macrophages thereby altering their mitochondrial metabolism through RIPK3. RIPK3 promotes mitochondrial fission leading to elevated oxidative stress via DRP1. This triggers MMP12 to lyse arterial matrix, thereby stimulating AAA. Administration of recombinant HMGB1 to WT, but not Ripk3-/- mice, recapitulates ALI-induced proteolytic collapse of arterial architecture. Deletion of RIPK3 in myeloid cells, DRP1 or MMP12 suppression in ALI-inflicted mice repress arterial stress and brake MMP12 release by transmural macrophages thereby maintaining a strengthened arterial framework refractory to AAA. Our results establish an inter-organ circuitry that alerts arterial macrophages to regulate vascular remodeling.


Assuntos
Lesão Pulmonar Aguda/complicações , Aneurisma da Aorta Abdominal/patologia , Proteína HMGB1/metabolismo , Macrófagos/metabolismo , Remodelação Vascular , Lesão Pulmonar Aguda/patologia , Animais , Aorta Abdominal/citologia , Aorta Abdominal/patologia , Aneurisma da Aorta Abdominal/etiologia , Aneurisma da Aorta Abdominal/prevenção & controle , Células Cultivadas , Modelos Animais de Doenças , Dinaminas/antagonistas & inibidores , Dinaminas/metabolismo , Humanos , Macrófagos/citologia , Metaloproteinase 12 da Matriz/genética , Metaloproteinase 12 da Matriz/metabolismo , Camundongos , Camundongos Knockout , Dinâmica Mitocondrial/efeitos dos fármacos , Estresse Oxidativo/efeitos dos fármacos , Fosforilação , Cultura Primária de Células , Proteólise/efeitos dos fármacos , Doença Pulmonar Obstrutiva Crônica/complicações , Doença Pulmonar Obstrutiva Crônica/patologia , Proteína Serina-Treonina Quinases de Interação com Receptores/genética , Proteína Serina-Treonina Quinases de Interação com Receptores/metabolismo , Estudos Retrospectivos , Regulação para Cima
2.
Proc Natl Acad Sci U S A ; 116(26): 13006-13015, 2019 06 25.
Artigo em Inglês | MEDLINE | ID: mdl-31189595

RESUMO

Abdominal aortic aneurysm (AAA) remains the second most frequent vascular disease with high mortality but has no approved medical therapy. We investigated the direct role of apelin (APLN) in AAA and identified a unique approach to enhance APLN action as a therapeutic intervention for this disease. Loss of APLN potentiated angiotensin II (Ang II)-induced AAA formation, aortic rupture, and reduced survival. Formation of AAA was driven by increased smooth muscle cell (SMC) apoptosis and oxidative stress in Apln-/y aorta and in APLN-deficient cultured murine and human aortic SMCs. Ang II-induced myogenic response and hypertension were greater in Apln-/y mice, however, an equivalent hypertension induced by phenylephrine, an α-adrenergic agonist, did not cause AAA or rupture in Apln-/y mice. We further identified Ang converting enzyme 2 (ACE2), the major negative regulator of the renin-Ang system (RAS), as an important target of APLN action in the vasculature. Using a combination of genetic, pharmacological, and modeling approaches, we identified neutral endopeptidase (NEP) that is up-regulated in human AAA tissue as a major enzyme that metabolizes and inactivates APLN-17 peptide. We designed and synthesized a potent APLN-17 analog, APLN-NMeLeu9-A2, that is resistant to NEP cleavage. This stable APLN analog ameliorated Ang II-mediated adverse aortic remodeling and AAA formation in an established model of AAA, high-fat diet (HFD) in Ldlr-/- mice. Our findings define a critical role of APLN in AAA formation through induction of ACE2 and protection of vascular SMCs, whereas stable APLN analogs provide an effective therapy for vascular diseases.


Assuntos
Aorta Abdominal/patologia , Aneurisma da Aorta Abdominal/patologia , Apelina/metabolismo , Neprilisina/metabolismo , Idoso , Idoso de 80 Anos ou mais , Angiotensina II/administração & dosagem , Enzima de Conversão de Angiotensina 2 , Animais , Aorta Abdominal/citologia , Aneurisma da Aorta Abdominal/tratamento farmacológico , Aneurisma da Aorta Abdominal/etiologia , Apelina/genética , Apoptose/efeitos dos fármacos , Apoptose/genética , Fármacos Cardiovasculares/química , Fármacos Cardiovasculares/farmacologia , Fármacos Cardiovasculares/uso terapêutico , Dieta Hiperlipídica/efeitos adversos , Modelos Animais de Doenças , Feminino , Técnicas de Silenciamento de Genes , Humanos , Masculino , Camundongos Transgênicos , Pessoa de Meia-Idade , Miócitos de Músculo Liso , Neprilisina/genética , Estresse Oxidativo/efeitos dos fármacos , Estresse Oxidativo/genética , Peptidil Dipeptidase A/metabolismo , Fenilefrina/administração & dosagem , Cultura Primária de Células , Proteólise/efeitos dos fármacos , RNA Interferente Pequeno/metabolismo , Receptores de LDL/genética , Receptores de LDL/metabolismo , Remodelação Vascular/efeitos dos fármacos , Remodelação Vascular/genética
3.
Mol Cell Biochem ; 459(1-2): 121-130, 2019 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-31127491

RESUMO

The antihyperglycemic agent empagliflozin not only improves glycemic control but has also been associated with clinically meaningful reductions in cardiovascular events. Studies have shown that empagliflozin significantly reduces cardiovascular death and heart failure-associated hospitalizations. Given that endothelial dysfunction is closely linked with the pathogenesis of atherosclerotic cardiovascular disease, we hypothesized that the cardiovascular benefits observed with empagliflozin may be a result of its positive impact on the health of the endothelial glycocalyx (GCX), a critical component for the endothelium homeostasis. Human abdominal aortic endothelial cells (HAAECs) were either statically cultured or subjected to a steady wall shear stress of 10 dyne/cm2. Empagliflozin (50 µM, 24 h) restored heparinase III-mediated GCX disruption and the normal mechanotransduction responses in GCX-compromised HAAECs while reducing the attachment of all-trans retinoic acid-transformed NB4 cells to HAAECs. The current body of work suggests that the cardioprotective properties previously reported for empagliflozin may in part be due to the ability of empagliflozin to preserve and restore the structural integrity of the GCX, which in turn helps to maintain vascular health by promoting an anti-inflammatory endothelium, in the presence of a pro-inflammatory environment. Further studies are needed to fully understand the mechanisms underlying the cardiovascular benefits of empagliflozin.


Assuntos
Aorta Abdominal/metabolismo , Compostos Benzidrílicos/farmacologia , Cardiotônicos/farmacologia , Células Endoteliais/metabolismo , Endotélio Vascular/metabolismo , Glucosídeos/farmacologia , Glicocálix/metabolismo , Aorta Abdominal/citologia , Linhagem Celular Tumoral , Células Endoteliais/citologia , Endotélio Vascular/citologia , Humanos
4.
J Oleo Sci ; 66(5): 499-506, 2017 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-28381776

RESUMO

Abdominal aortic aneurysm (AAA) is a vascular disease that results in the gradual dilation of the abdominal aorta and has a high rupture-related mortality rate. However, the mechanism of AAA rupture remains unknown. In our previous study, we established a novel AAA animal model (hypoperfusion-induced AAA rat model) with spontaneous AAA rupture. Using the hypoperfusion-induced AAA rat model, we demonstrated that the abnormal appearance of adipocytes in the vascular wall is associated with AAA rupture. However, pathological analysis of the rupture area has not been performed because it is particularly difficult to identify the rupture point. In this study, we succeeded in obtaining samples from the rupture point and performed a histological analysis of the ruptured area in the vascular wall in the hypoperfusion-induced AAA rat model. Adipocytes were observed along the AAA-ruptured area of the vascular wall. In the areas around the adipocytes, macrophage infiltration and protein levels of matrix metalloproteinases 2 and 9 were significantly increased and collagen-positive areas were significantly decreased, as compared with areas without adipocytes. The AAA diameter was correlated with the number of adipocytes in the vascular wall of the hypoperfusion-induced AAA rat model. On the other hand, serum triglyceride levels and serum total cholesterol levels were not correlated with the number of adipocytes in the vascular wall. These results suggest that local adipocyte accumulation in the vascular wall, not serum lipids, has an important role in AAA rupture.


Assuntos
Adipócitos/patologia , Aorta Abdominal/patologia , Aneurisma da Aorta Abdominal/patologia , Ruptura Aórtica/patologia , Modelos Animais de Doenças , Adipócitos/metabolismo , Animais , Aorta Abdominal/citologia , Aneurisma da Aorta Abdominal/etiologia , Aneurisma da Aorta Abdominal/metabolismo , Ruptura Aórtica/etiologia , Ruptura Aórtica/metabolismo , Colágeno/metabolismo , Macrófagos/patologia , Masculino , Metaloproteinase 2 da Matriz/metabolismo , Metaloproteinase 9 da Matriz/metabolismo , Ratos Sprague-Dawley
5.
Cell Transplant ; 26(2): 173-189, 2017 02 16.
Artigo em Inglês | MEDLINE | ID: mdl-27436185

RESUMO

Abdominal aortic aneurysm (AAA) is a potentially lethal disease associated with immune activation-induced aortic degradation. We hypothesized that xenotransplantation of human adipose-derived stem cells (hADSCs) would reduce aortic inflammation and attenuate expansion in a murine AAA model. Modulatory effects of ADSCs on immune cell subtypes associated with AAA progression were investigated using human peripheral blood mononuclear cells (hPBMNCs) cocultured with ADSCs. Murine AAA was induced through elastase application to the abdominal aorta in C57BL/6 mice. ADSCs were administered intravenously, and aortic changes were determined by ultrasonography and videomicrometry. Circulating monocytes, aortic neutrophils, CD28- T cells, FoxP3+ regulatory T cells (Tregs), and CD206+ M2 macrophages were assessed at multiple terminal time points. In vitro, ADSCs induced M2 macrophage and Treg phenotypes while inhibiting neutrophil transmigration and lymphocyte activation without cellular contact. Intravenous ADSC delivery reduced aneurysmal expansion starting from day 4 [from baseline: 54.8% (saline) vs. 16.9% (ADSCs), n = 10 at baseline, n = 4 at day 4, p < 0.001], and the therapeutic effect persists through day 14 (from baseline: 64.1% saline vs. 24.6% ADSCs, n = 4, p < 0.01). ADSC administration increased aortic Tregs by 20-fold (n = 5, p < 0.01), while decreasing CD4+CD28- (-28%), CD8+CD28- T cells (-61%), and Ly6G/C+ neutrophils (-43%, n = 5, p < 0.05). Circulating CD115+CXCR1-LY6C+-activated monocytes decreased in the ADSC-treated group by day 7 (-60%, n = 10, p < 0.05), paralleled by an increase in aortic CD206+ M2 macrophages by 2.4-fold (n = 5, p < 0.05). Intravenously injected ADSCs transiently engrafted in the lung on day 1 without aortic engraftment at any time point. In conclusion, ADSCs exhibit pleiotropic immunomodulatory effects in vitro as well as in vivo during the development of AAA. The temporal evolution of these effects systemically as well as in aortic tissue suggests that ADSCs induce a sequence of anti-inflammatory cellular events mediated by paracrine factors, which leads to amelioration of AAA progression.


Assuntos
Aorta Abdominal/citologia , Aneurisma da Aorta Abdominal/metabolismo , Macrófagos/metabolismo , Elastase Pancreática/metabolismo , Células-Tronco/citologia , Animais , Aneurisma da Aorta Abdominal/enzimologia , Aneurisma da Aorta Abdominal/imunologia , Células Cultivadas , Citometria de Fluxo , Humanos , Imuno-Histoquímica , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Microscopia de Vídeo , Reação em Cadeia da Polimerase em Tempo Real , Células-Tronco/fisiologia , Linfócitos T Reguladores/metabolismo
6.
Clin Exp Pharmacol Physiol ; 42(5): 510-9, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25740656

RESUMO

Microgravity-induced vascular remodelling may play an important role in post-spaceflight orthostatic intolerance. In this study, we aimed to investigate the effects of simulated microgravity on monocyte adhesion to aortic endothelium in hindlimb unweighted rats and to elucidate the underlying mechanisms associated with this event. Sprague-Dawley rats were subjected to 4-week hindlimb unweighting to simulate microgravity. The recruitment of monocytes to the abdominal aorta was investigated by en face immunofluorescence staining and monocyte binding assays. The expression of the adhesion molecules E-selectin and vascular cell adhesion molecule-1 as well as the cytokine monocyte chemoattractant protein (MCP)-1 was evaluated by immunohistochemical staining, western blot, and quantitative reverse transcription polymerase chain reaction analyses. Additionally, nuclear factor-κB (NF-κB) activation and the messenger RNA expression levels of E-selectin, vascular cell adhesion molecule-1, and MCP-1 were assessed with the administration of an NF-κB inhibitor, pyrrolidine dithiocarbamate. Results showed that simulated microgravity significantly increased monocyte recruitment to the aortic endothelium, protein expression of E-selectin and MCP-1, and NF-κB activation in the abdominal aorta of rats. Pyrrolidine dithiocarbamate treatment not only significantly inhibited NF-κB activity but also reduced the messenger RNA levels of E-selectin, vascular cell adhesion molecule-1, and MCP-1 as well as monocyte recruitment in the abdominal aorta of hindlimb unweighted rats. These results suggest that simulated microgravity increases monocyte adhesion to rat aortic endothelium via the NF-κB-mediated expression of the adhesion molecule E-selectin and the cytokine MCP-1. Therefore, an NF-κB-mediated inflammatory response may be one of the cellular mechanisms responsible for arterial remodelling during exposure to microgravity.


Assuntos
Aorta Abdominal/citologia , Endotélio Vascular/citologia , Monócitos/citologia , NF-kappa B/metabolismo , Simulação de Ausência de Peso , Transporte Ativo do Núcleo Celular/efeitos dos fármacos , Animais , Adesão Celular/efeitos dos fármacos , Núcleo Celular/efeitos dos fármacos , Núcleo Celular/metabolismo , Quimiocina CCL2/genética , Selectina E/genética , Endotélio Vascular/efeitos dos fármacos , Regulação da Expressão Gênica/efeitos dos fármacos , Macrófagos/citologia , Macrófagos/efeitos dos fármacos , Masculino , Monócitos/efeitos dos fármacos , NF-kappa B/antagonistas & inibidores , Pirrolidinas/farmacologia , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Ratos , Ratos Sprague-Dawley , Tiocarbamatos/farmacologia , Molécula 1 de Adesão de Célula Vascular/genética
7.
Biochem Biophys Res Commun ; 444(2): 205-11, 2014 Feb 07.
Artigo em Inglês | MEDLINE | ID: mdl-24440697

RESUMO

Abdominal aortic aneurysms (AAA) are progressive dilatations of infra-renal aorta causing structural weakening rendering the aorta prone to rupture. AAA can be potentially stabilized by inhibiting inflammatory enzymes such as matrix metalloproteinases (MMP); however, active regression of AAA is not possible without new elastic fiber regeneration. Here we report the elastogenic benefit of direct delivery of polyphenols such as pentagalloyl glucose (PGG), epigallocatechin gallate (EGCG), and catechin, to smooth muscle cells obtained either from healthy or from aneurysmal rat aorta. Addition of 10 µg/ml PGG and ECGC induce elastin synthesis, organization, and crosslinking while catechin does not. Our results indicate that polyphenols bind to monomeric tropoelastin and enhance coacervation, aid in crosslinking of elastin by increasing lysyl oxidase (LOX) synthesis, and by blocking MMP-2 activity. Thus, polyphenol treatments leads to increased mature elastin fibers synthesis without increasing the production of intracellular tropoelastin.


Assuntos
Tecido Elástico/metabolismo , Miócitos de Músculo Liso/efeitos dos fármacos , Polifenóis/farmacologia , Tropoelastina/metabolismo , Animais , Aorta Abdominal/citologia , Aorta Abdominal/metabolismo , Aorta Abdominal/fisiologia , Aneurisma da Aorta Abdominal/metabolismo , Aneurisma da Aorta Abdominal/patologia , Aneurisma da Aorta Abdominal/fisiopatologia , Catequina/análogos & derivados , Catequina/metabolismo , Catequina/farmacologia , Células Cultivadas , Tecido Elástico/ultraestrutura , Elastina/metabolismo , Elastina/ultraestrutura , Fibrilinas , Taninos Hidrolisáveis/metabolismo , Taninos Hidrolisáveis/farmacologia , Cinética , Masculino , Metaloproteinase 2 da Matriz/metabolismo , Proteínas dos Microfilamentos/metabolismo , Microscopia Eletrônica de Transmissão , Microscopia de Fluorescência , Músculo Liso Vascular/citologia , Miócitos de Músculo Liso/metabolismo , Miócitos de Músculo Liso/ultraestrutura , Polifenóis/metabolismo , Proteína-Lisina 6-Oxidase/metabolismo , Ratos , Ratos Sprague-Dawley , Regeneração
8.
Fundam Clin Pharmacol ; 27(5): 465-70, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22607657

RESUMO

Regulation of angiogenesis involves tight cell-to-cell and cell-to-extracellular-matrix interactions. Various reports demonstrate that the Wnt signaling pathways participate in this regulation. Using a three-dimensional aortic ring culture combined with an ex vivo retroviral infection approach, we evaluated the effects of two Wnt growth factors, Wnt-1 and Wnt-4, on the formation and growth of new capillaries. Our results show that Wnt-1 had no effect, whereas Wnt-4 was a potent inhibitor of capillary outgrowth in vitro.


Assuntos
Inibidores da Angiogênese/metabolismo , Aorta Abdominal/metabolismo , Capilares/metabolismo , Endotélio Vascular/metabolismo , Matriz Extracelular/metabolismo , Neovascularização Fisiológica , Proteína Wnt4/metabolismo , Células 3T3 , Inibidores da Angiogênese/genética , Animais , Aorta Abdominal/citologia , Capilares/citologia , Técnicas de Cocultura , Endotélio Vascular/citologia , Glicoproteínas/genética , Glicoproteínas/metabolismo , Imageamento Tridimensional , Imuno-Histoquímica , Peptídeos e Proteínas de Sinalização Intracelular , Masculino , Camundongos , Ratos , Ratos Sprague-Dawley , Proteínas Recombinantes/metabolismo , Técnicas de Cultura de Tecidos , Proteína Wnt1/genética , Proteína Wnt1/metabolismo , Proteína Wnt4/genética
9.
Cardiovasc Res ; 97(3): 544-52, 2013 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-23250921

RESUMO

AIMS: Macrophages (MPs) and vascular smooth muscle cells (VSMCs) closely interact within the growing atherosclerotic plaque. An in vitro co-culture model was established to study how MPs modulate VSMC behaviour. METHODS AND RESULTS: MPs were exposed to fluorescence-labelled-acetylated LDL (FL-acLDL) prior to co-culture with VSMCs. Fluorescence microscopy visualized first transport of FL-acLDL within 6 h after co-culture implementation. When MPs had been fed with FL-acLDL in complex with fluorescence-labelled cholesterol (FL-Chol), these complexes were also transferred during co-culture and resulted in cholesterol positive lipid droplet formation in VSMCs. When infected with a virus coding for a fusion protein of Rab5a and fluorescent protein reporter (FP) to mark early endosomes, no co-localization between Rab5a-FP and the transported FL-acLDL within VSMCs was detected implying a mechanism independent of phagocytosis. Next, expression of lysosome-associated membrane glycoprotein 1 (LAMP1)-FP, marking all lysosomes in VSMCs, revealed that the FL-acLDL was located in non-acidic lysosomes. MPs infected with virus encoding for LAMP1-FP prior to co-culture demonstrated that intact fluorescence-marked lysosomes were transported into the VSMC, instead. Xenogenic cell composition (rat VSMC, human MP) and subsequent quantitative RT-PCR with rat-specific primers rendered induction of genes typical for MPs and down-regulation of the cholesterol sensitive HMG-CoA reductase. CONCLUSION: Our results demonstrate that acLDL/cholesterol-loaded lysosomes are transported from MPs into VSMCs in vitro. Lysosomal transfer results in a phenotypic alteration of the VSMC towards a foam cell-like cell. This way VSMCs may lose their plaque stabilizing properties and rather contribute to plaque destabilization and rupture.


Assuntos
Colesterol/metabolismo , Lipoproteínas LDL/metabolismo , Lisossomos/metabolismo , Macrófagos/metabolismo , Músculo Liso Vascular/metabolismo , Fenótipo , Animais , Aorta Abdominal/citologia , Aorta Abdominal/metabolismo , Comunicação Celular/fisiologia , Células Cultivadas , LDL-Colesterol/metabolismo , Técnicas de Cocultura , Humanos , Hidroximetilglutaril-CoA Redutases/metabolismo , Proteína 1 de Membrana Associada ao Lisossomo/metabolismo , Macrófagos/citologia , Microscopia de Fluorescência , Músculo Liso Vascular/citologia , Ratos , Ratos Wistar
10.
Proc Natl Acad Sci U S A ; 109(20): 7770-5, 2012 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-22550179

RESUMO

Vascular endothelial cells (ECs) are constantly exposed to blood flow-induced shear stress, but the mechanism of force-specific activation of their signaling to modulate cellular function remains unclear. We have demonstrated that bone morphogenetic protein receptor (BMPR)-specific Smad1/5 can be force-specifically activated by oscillatory shear stress (OSS) in ECs to cause cell cycle progression. Smad1/5 is highly activated in ECs of atherosclerotic lesions in diseased human coronary arteries from patients with end-stage heart failure undergoing heart transplantation and from apolipoprotein E-deficient mice. Application of OSS (0.5 ± 4 dyn/cm(2)) causes the sustained activation of Smad1/5 in ECs through activations of mammalian target of rapamycin and p70S6 kinase, leading to up-regulation of cyclin A and down-regulations of p21(CIP1) and p27(KIP1) and, hence, EC cycle progression. En face examination of rat aortas reveals high levels of phospho-Smad1/5 in ECs of the inner, but not the outer, curvature of aortic arch, nor the straight segment of thoracic aorta [corrected]. Immunohistochemical and en face examinations of the experimentally stenosed abdominal aorta in rats show high levels of phospho-Smad1/5 in ECs at poststenotic sites, where OSS occurs. These OSS activations of EC Smad1/5 in vitro and in vivo are not inhibited by the BMP-specific antagonist Noggin and, hence, are independent of BMP ligand. Transfecting ECs with Smad1/5-specific small interfering RNAs inhibits the OSS-induced EC cycle progression. Our findings demonstrate the force-specificity of the activation of Smad1/5 and its contribution to cell cycle progression in ECs induced by disturbed flow.


Assuntos
Aterosclerose/fisiopatologia , Ciclo Celular/fisiologia , Células Endoteliais/fisiologia , Regulação da Expressão Gênica/fisiologia , Fluxo Sanguíneo Regional/fisiologia , Proteína Smad1/metabolismo , Estresse Mecânico , Animais , Aorta Abdominal/citologia , Aorta Abdominal/patologia , Apolipoproteínas E/genética , Fenômenos Biomecânicos , Vasos Coronários/citologia , Vasos Coronários/patologia , Ciclina A/metabolismo , Inibidor de Quinase Dependente de Ciclina p21/metabolismo , Inibidor de Quinase Dependente de Ciclina p27/metabolismo , Humanos , Imuno-Histoquímica , Camundongos , Camundongos Knockout , Ratos , Proteínas Quinases S6 Ribossômicas 70-kDa/metabolismo , Serina-Treonina Quinases TOR/metabolismo , Taiwan
11.
J Mater Sci Mater Med ; 23(5): 1235-45, 2012 May.
Artigo em Inglês | MEDLINE | ID: mdl-22359212

RESUMO

A novel 316L type Cu-bearing stainless steel was developed in present work, aiming at reducing the occurrence of the in-stent restenosis after implantations of coronary stents, through trace amount of Cu release from surface of the steel in body fluid. It was found that there was a trace amount of Cu released from the Cu-bearing steel in a simulated body fluid, with no cytotoxicity. All the in vitro experimental results proved that this Cu-bearing steel could not only inhibit the proliferation of vascular smooth muscle cells, reducing the formation of thrombosis, which are the main reasons for happening of the in-stent restenosis, but also promote the proliferation of vascular endothelial cells needed for the revascularization, showing that this novel steel is prospective to be a new material for manufacturing coronary stents with function of reducing the in-stent restenosis.


Assuntos
Prótese Vascular , Cobre/farmacocinética , Oclusão de Enxerto Vascular/prevenção & controle , Aço Inoxidável/farmacocinética , Stents , Animais , Aorta Abdominal/citologia , Líquidos Corporais/química , Líquidos Corporais/metabolismo , Células Cultivadas , Cobre/análise , Cobre/química , Cobre/fisiologia , Desenho de Equipamento , Análise de Falha de Equipamento , Oclusão de Enxerto Vascular/etiologia , Oclusão de Enxerto Vascular/metabolismo , Humanos , Microtecnologia/métodos , Músculo Liso Vascular/citologia , Músculo Liso Vascular/efeitos dos fármacos , Músculo Liso Vascular/metabolismo , Músculo Liso Vascular/fisiologia , Miócitos de Músculo Liso/citologia , Miócitos de Músculo Liso/efeitos dos fármacos , Miócitos de Músculo Liso/metabolismo , Miócitos de Músculo Liso/fisiologia , Concentração Osmolar , Ratos , Aço Inoxidável/química , Tempo de Coagulação do Sangue Total
12.
Neurosci Lett ; 499(2): 104-8, 2011 Jul 20.
Artigo em Inglês | MEDLINE | ID: mdl-21651959

RESUMO

The purpose of this study was to investigate the effects of a decellularized artery allograft containing autologous adipose-derived stem cells (ADSCs) on an 8-mm facial nerve branch lesion in a rat model. At 8 weeks postoperatively, functional evaluation of unilateral vibrissae movements, morphological analysis of regenerated nerve segments and retrograde labeling of facial motoneurons were all analyzed. Better regenerative outcomes associated with functional improvement, great axonal growth, and improved target reinnervation were achieved in the artery-ADSCs group (2), whereas the cut nerves sutured with artery conduits alone (group 1) achieved inferior restoration. Furthermore, transected nerves repaired with nerve autografts (group 3) resulted in significant recovery of whisking, maturation of myelinated fibers and increased number of labeled facial neurons, and the latter two parameters were significantly different from those of group 2. Collectively, though our combined use of a decellularized artery allograft with autologous ADSCs achieved regenerative outcomes inferior to a nerve autograft, it certainly showed a beneficial effect on promoting nerve regeneration and thus represents an alternative approach for the reconstruction of peripheral facial nerve defects.


Assuntos
Gordura Abdominal/transplante , Aorta Abdominal/transplante , Modelos Animais de Doenças , Traumatismos do Nervo Facial/cirurgia , Transplante de Células-Tronco/métodos , Gordura Abdominal/citologia , Gordura Abdominal/fisiologia , Adipócitos/citologia , Adipócitos/fisiologia , Adipócitos/transplante , Animais , Aorta Abdominal/citologia , Traumatismos do Nervo Facial/patologia , Feminino , Regeneração Nervosa/fisiologia , Distribuição Aleatória , Ratos , Ratos Sprague-Dawley , Transplante Autólogo , Vibrissas/inervação , Vibrissas/fisiologia
13.
Stem Cell Rev Rep ; 7(4): 847-59, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-21472453

RESUMO

Engineering living, multilayered blood vessels to form in vivo arteries is a promising alternative to peripheral artery bypass using acellular grafts restricted by thrombosis and occlusion at long term. Bone Morphogenetic Protein 2 (BMP2) is a growth factor determining in the early vascular embryonic development. The aim of the present study was evaluate the collaborative effect of recombinant human--BMP2 and Bone marrow--Mesenchymal stem cells (BM-MSCs) seeded on vascular patch to regenerate a vascular arterial wall in a rat model. BM-MSCs expressing green fluorescent protein (GFP) seeded on vascular patch were cultured in presence of recombinant human-BMP2 [100 ng/mL] during 1 week before their implantation on the abdominal aorta of Wistar rats. We observed after 2 weeks under physiological arterial flow a regeneration of a three layers adult-like arterial wall with a middle layer expressing smooth muscle proteins and a border layer expressing endothelial marker. In vitro study, using Matrigel assay and co-culture of BM-MSCs with endothelial cells demonstrated that rh-BMP2 promoted tube-like formation even at long term (90 days) allowing the organization of thick rails. We demonstrated using inhibitors and siRNAs that rh-BMP2 enhanced the expression of HIF-1α and Id1 through, at least in part, the stimulation of JAK2/STAT3/STAT5 signaling pathways. Rh-BMP2 by mimicking embryological conditions allowed vascular BM-MSCs differentiation.


Assuntos
Proteína Morfogenética Óssea 2/farmacologia , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Proteína 1 Inibidora de Diferenciação/metabolismo , Células-Tronco Mesenquimais/citologia , Regeneração , Animais , Aorta Abdominal/citologia , Aorta Abdominal/metabolismo , Aorta Abdominal/transplante , Biomarcadores/metabolismo , Prótese Vascular , Diferenciação Celular , Técnicas de Cocultura , Proteínas do Citoesqueleto/metabolismo , Células Endoteliais/citologia , Células Endoteliais/metabolismo , Proteínas de Fluorescência Verde/metabolismo , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Proteína 1 Inibidora de Diferenciação/genética , Janus Quinases/genética , Janus Quinases/metabolismo , Masculino , Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais/efeitos dos fármacos , Modelos Animais , Proteínas Musculares/metabolismo , Neovascularização Fisiológica , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Ratos , Ratos Wistar , Proteínas Recombinantes/farmacologia , Transdução de Sinais , Fatores de Tempo , Transfecção/métodos
14.
Microsurgery ; 31(2): 138-45, 2011 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-21268111

RESUMO

The objective of this preliminary study was to develop a reabsorbable vascular patch that did not require in vitro cell or biochemical preconditioning for vascular wall repair. Patches were composed only of hyaluronic acid (HA). Twenty male Wistar rats weighing 250-350 g were used. The abdominal aorta was exposed and isolated. A rectangular breach (1 mm × 5 mm) was made on vessel wall and arterial defect was repaired with HA made patch. Performance was assessed at 1, 2, 4, 8, and 16 weeks after surgery by histology and immunohistochemistry. Extracellular matrix components were evaluated by molecular biological methods. After 16 weeks, the biomaterial was almost completely degraded and replaced by a neoartery wall composed of endothelial cells, smooth muscle cells, collagen, and elastin fibers organized in layers. In conclusion, HA patches provide a provisional three-dimensional support to interact with cells for the control of their function, guiding the spatially and temporally multicellular processes of artery regeneration.


Assuntos
Implantes Absorvíveis , Regeneração Tecidual Guiada/instrumentação , Ácido Hialurônico , Alicerces Teciduais , Animais , Aorta Abdominal/citologia , Aorta Abdominal/lesões , Aorta Abdominal/fisiologia , Aorta Abdominal/cirurgia , Biomarcadores/análise , Expressão Gênica , Regeneração Tecidual Guiada/métodos , Masculino , Projetos Piloto , Ratos , Ratos Wistar , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Resultado do Tratamento , Cicatrização
15.
Acta Pharmacol Sin ; 31(4): 413-20, 2010 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-20348945

RESUMO

AIM: To investigate whether adenosine A(3) receptors (A(3)AR) stimulation restore vascular reactivity after hemorrhagic shock through a ryanodine receptor (RyR)-mediated and large conductance calcium-activated potassium (BK(Ca)) channel-dependent pathway. METHODS: Rat hemorrhagic shock model (40 mmHg) and vascular smooth muscle cell (VSMC) hypoxic model were used. The expression of A(3)AR was determined by Western blot and RT-PCR. The effect of A(3)AR stimulation on RyR-mediated Ca(2+) release in VSMCs was analyzed by the Fura-3/AM loading Ca(2+) imaging. The modulation of vascular reactivity to norepinephrine (NE) by A(3)AR stimulation was monitored by an isolated organ tension instrument. RESULTS: Decrease of A(3)AR expression is consistent with the loss of vasoreactivity to NE in hemorrhagic shock rats. The stimulation of A(3)AR with a selective agonist, IB-MECA, could partly but significantly restore the vasoreactivity in the rats, and this restorative effect could be counteracted by MRS1523, a selective A(3)AR antagonist. In hypoxic VSMCs, RyR activation by caffeine significantly evoked the rise of [Ca(2+)] compared with the control cells, a phenomenon closely associated with the development of vascular hyporeactivity in hemorrhagic shock rats. The stimulation of A(3)AR with IB-MECA significantly blocked this over activation of RyR-mediated Ca(2+) release. RyR activation by caffeine and BK(Ca) channel activation by NS1619 attenuated the restoration of vasoreactivity to NE resulting from A(3)AR stimulation by IB-MECA after hemorrhagic shock; this attenuation effect could be antagonized by a selective BK(Ca) channel blocker. CONCLUSION: These findings suggest that A(3)AR is involved in the modulation of vasoreactivity after hemorrhagic shock and that stimulation of A(3)AR can restore the decreased vasoreactivity to NE through a RyR-mediated, BK(Ca) channel-dependent signal pathway.


Assuntos
Canais de Potássio Ativados por Cálcio de Condutância Alta/metabolismo , Músculo Liso Vascular/efeitos dos fármacos , Norepinefrina/farmacologia , Receptor A3 de Adenosina/metabolismo , Choque Hemorrágico/metabolismo , Vasoconstritores/farmacologia , Animais , Aorta Abdominal/citologia , Cálcio/metabolismo , Hipóxia Celular/efeitos dos fármacos , Células Cultivadas , Expressão Gênica , Músculo Liso Vascular/citologia , Ratos , Ratos Sprague-Dawley , Ratos Wistar , Receptor A3 de Adenosina/genética , Canal de Liberação de Cálcio do Receptor de Rianodina/metabolismo , Vasoconstrição/efeitos dos fármacos
16.
J Vasc Surg ; 51(5): 1248-59, 2010 May.
Artigo em Inglês | MEDLINE | ID: mdl-20223627

RESUMO

OBJECTIVE: Isopropylamine NONOate (IPA/NO) is a nitroxyl (HNO) donor at physiologic pH. HNO is a positive inotrope and vasodilator, but little is known about its effect on neointimal hyperplasia. The aims of this study are to determine the effect of IPA/NO on endothelial and vascular smooth muscle cells (VSMC) in vitro and to determine if IPA/NO inhibits neointimal hyperplasia in vivo. METHODS: VSMC were harvested from the abdominal aortas of male Sprague Dawley rats, and human umbilical vein endothelial cells were purchased from ATCC. In vitro, cellular proliferation was assessed by (3)H-thymidine incorporation, cell migration was assessed using the scrape assay, and cell death was assessed using Guava personal cell analysis (PCA). Cell cycle analysis was performed using propidium iodide staining and flow cytometry analysis. Protein expression was assessed using Western blot analysis. Phosphorylated proteins were assessed using immunoprecipitation and Western blot analysis. In vivo, the carotid artery injury model was performed on male Sprague Dawley rats treated with (n = 12) or without (n = 6) periadventitial IPA/NO (10 mg). Arteries harvested at 2 weeks were assessed for morphometrics using ImageJ. Inflammation was assessed using immunohistochemistry. Endothelialization was assessed by Evans blue staining of carotid arteries harvested 7 days after balloon injury from rats treated with (n = 6) or without (n = 3) periadventitial IPA/NO (10 mg). RESULTS: In vitro, 1000 micromol/L IPA/NO inhibited both VSMC (38.7 +/- 4.5% inhibition vs control, P = .003) and endothelial cell proliferation (54.0 +/- 2.9% inhibition vs control, P < or = 0.001) without inducing cell death or inhibiting migration. In VSMC, this inhibition was associated with an S-phase cell cycle arrest and increased expression of cyclin A, cyclin D1, and the cyclin-dependent kinase inhibitor p21. No change was noted in the phosphorylation status of cdk2, cdk4, or cdk6 by IPA/NO. In rodents subjected to the carotid artery balloon injury model, IPA/NO caused significant reductions in neointimal area (298 +/- 20 vs 422 +/- 30, P < or = .001) and medial area (311 +/- 14 vs 449 +/- 16, P < or = .001) compared with injury alone, and reduced macrophage infiltration to 1.7 +/- 0.8 from 16.1 +/- 3.5 cells per high power field (P < or = .001). IPA/NO also prevented re-endothelialization compared with injury alone (55.9 +/- 0.5% nonendothelialized vs 21 +/- 4.4%, respectively, P = .001). Lastly, a 50% mortality rate was observed in the IPA/NO-treated groups. CONCLUSIONS: In summary, while IPA/NO modestly inhibited neointimal hyperplasia by inhibiting VSMC proliferation and macrophage infiltration, it also inhibited endothelial cell proliferation and induced significant mortality in our animal model. Since HNO is being investigated as a treatment for congestive heart failure, our results raise some concerns about the use of IPA/NO in the vasculature and suggest that further studies be conducted on the safety of HNO donors in the cardiovascular system.


Assuntos
Lesões das Artérias Carótidas/metabolismo , Células Endoteliais/metabolismo , Células Endoteliais/patologia , Hidrazinas/metabolismo , Músculo Liso Vascular/metabolismo , Túnica Íntima/metabolismo , Túnica Íntima/patologia , Animais , Aorta Abdominal/citologia , Aorta Abdominal/efeitos dos fármacos , Western Blotting , Lesões das Artérias Carótidas/complicações , Lesões das Artérias Carótidas/patologia , Morte Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Modelos Animais de Doenças , Células Endoteliais/efeitos dos fármacos , Humanos , Hidrazinas/administração & dosagem , Hiperplasia/etiologia , Hiperplasia/metabolismo , Hiperplasia/patologia , Hiperplasia/prevenção & controle , Imuno-Histoquímica , Masculino , Músculo Liso Vascular/efeitos dos fármacos , Distribuição Aleatória , Ratos , Ratos Sprague-Dawley , Valores de Referência , Túnica Íntima/efeitos dos fármacos , Veias Umbilicais/citologia , Veias Umbilicais/efeitos dos fármacos
17.
Int J Cardiol ; 138(1): 70-80, 2010 Jan 07.
Artigo em Inglês | MEDLINE | ID: mdl-19095317

RESUMO

BACKGROUND: Endothelial repair is one of key events after vascular injury. The mechanisms by which hepatocyte growth factor (HGF) and endothelial progenitor cells (EPCs) may be responsible for re-endothelialization of injured blood vessel wall are poorly understood. METHODS: Primary culture SMCs were transfected with pcDNA3.0-HGF followed by G418 selection, one of G418-resistant colonies in well was picked, propagated and used as donor cells for further experiments. HGF and VEGF expression in SMCs were detected with western blot and enzyme linked immunosorbent assays (ELISA). Rat EPCs were cultured in untreated, pcDNA3.0 and pcDNA3.0-HGF transfected SMCs conditioned medium with or without anti-VEGF or exogenous recombinant HGF addition. eNOS, KDR and CD31 expression in EPCs was determined by real-time quantitative polymerase chain reaction (RT-qPCR) or flow cytometry; EPCs migration and proliferation were measured by using a modified Boyden chambers and MTT assay respectively. RESULTS: Abundant and stable expression of HGF was found in G418-resistant colony-derived SMCs. VEGF expression significantly increased in HGF transfected SMCs. Exogenous recombinant HGF (rHGF) markedly up-regulated eNOS mRNA expression in EPCs and promoted EPCs migration and proliferation, but no significant changes were found in KDR and CD31 mRNA expression. HGF transfection in SMCs was more effective than exogenous HGF for EPCs differentiation, proliferation and migration. CONCLUSIONS: Over-expression of HGF in SMCs can be helpful for promoting EPCs differentiation, increasing EPCs migration and proliferation. It may be responsible for angiogenesis of arteriosclerosis lesions and useful for blood vessel tissue engineering.


Assuntos
Movimento Celular/fisiologia , Células Endoteliais/citologia , Células-Tronco Hematopoéticas/citologia , Fator de Crescimento de Hepatócito/genética , Músculo Liso Vascular/fisiologia , Animais , Aorta Abdominal/citologia , Células da Medula Óssea/citologia , Células da Medula Óssea/fisiologia , Diferenciação Celular/fisiologia , Divisão Celular/fisiologia , Células Cultivadas , Citometria de Fluxo , Expressão Gênica/fisiologia , Células-Tronco Hematopoéticas/fisiologia , Fator de Crescimento de Hepatócito/metabolismo , Masculino , Músculo Liso Vascular/citologia , Ratos , Ratos Sprague-Dawley , Transfecção , Fator A de Crescimento do Endotélio Vascular/genética , Fator A de Crescimento do Endotélio Vascular/metabolismo
18.
Sichuan Da Xue Xue Bao Yi Xue Ban ; 41(5): 784-8, 2010 Sep.
Artigo em Chinês | MEDLINE | ID: mdl-21302441

RESUMO

OBJECTIVE: To investigate the effects of high glucose on expression of core binding factor alpha1 (cbfalpha-1) and osteocalcin (OC) in vascular smooth muscle cells (VSMCs), and discuss the mechanism of small vessels calcification induced by high glucose (GS) in vitro. METHODS: The primary cultured VSMCs from rats' aortic segments were divided into three groups, including normal control group (5 mmol/L D-glucose), high glucose group (25 mmol/L D-glucose) and mannitol group (5 mmol/L D-glucose plus 25 mmol/L mannitol). We measured quantitatively the calcium deposition in VSMCs and investigated the calcium extent of VSMCs by alizarin red stain in each group. The mRNA levels of cbfalpha-1 and OC were measured by real-time PCR, and the protein expression levels of cbfalpha-1 and OC were examined by Western blot. The activity of alkaline phosphatase was measured by alkaline phosphatase activity testing kit, and the protein level of alpha-smooth muscle actin (a-SMA) was detected by immunohistochemistry. RESULTS: When compared with the normal group and mannitol group, the high glucose group showed that the calcium deposition and calcium extent of VSMCs increased obviously, the mRNA and protein levels of cbfalpha-1 and OC also increased significantly (P < 0.05), while the protein level of alpha-SMA decreased (P < 0.05), which were in a dose-dependent manner. The level of alkaline phosphatase activity of VSMCs was approximately doubled in high glucose group. CONCLUSION: The mechanism of high glucose induced calcification in VSMCs may be due to the increased expression of cbfalpha-1 and OC. High glucose decrease the expression of alpha-SMA in VSMCs, which could induce the transdifferentiation from RVSMCs to osteoblast-like cells.


Assuntos
Subunidade alfa 1 de Fator de Ligação ao Core/metabolismo , Glucose/farmacologia , Músculo Liso Vascular/metabolismo , Osteocalcina/metabolismo , Animais , Aorta Abdominal/citologia , Células Cultivadas , Subunidade alfa 1 de Fator de Ligação ao Core/genética , Músculo Liso Vascular/citologia , Osteocalcina/genética , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Ratos , Ratos Sprague-Dawley
19.
J Thorac Cardiovasc Surg ; 138(6): 1392-9, 2009 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19931668

RESUMO

OBJECTIVES: Vascular smooth muscle cells can undergo profound changes in phenotype, defined by coordinated repression of smooth muscle cell marker genes and production of matrix metalloproteinases in response to injury. However, little is known of the role of smooth muscle cells in aortic aneurysms. We hypothesized that smooth muscle cells undergo phenotypic modulation early in the development of aortic aneurysms. METHODS: Abdominal aortas from C57B6 mice (n = 79) were perfused with elastase or saline (control) and harvested at 1, 3, 7, or 14 days. Aortas were analyzed by means of quantitative polymerase chain reaction and immunohistochemistry for smooth muscle cell marker genes, including SM22A, smooth muscle alpha-actin, and matrix metalloproteinases 2 and 9. In complimentary experiments human aneurysms (n = 10) and control aorta (n = 10) were harvested at the time of surgical intervention and analyzed. RESULTS: By 14 days, aortic diameter was larger after elastase perfusion compared with control diameter (100% +/- 9.6% vs 59.5% +/- 18.9%, P = .0002). At 7 days, elastase-perfused mice had a 78% and 85% reduction in SM22 alpha and smooth muscle alpha-actin expression, respectively, compared with that seen in control animals well before aneurysms were present, and these values remained repressed at 14 days. Immunohistochemistry confirmed less SM22 alpha and smooth muscle alpha-actin in experimental aneurysms at 14 days in concert with increased matrix metalloproteinase 2 and 9 expression at 7 and 14 days. Similarly, human aneurysms had less SM22 alpha and smooth muscle alpha-actin and increased matrix metalloproteinase 2 and 9 staining, compared with control values, as determined by means of quantitative polymerase chain reaction. CONCLUSIONS: Aneurysms demonstrate smooth muscle cell phenotypic modulation characterized by downregulation of smooth muscle cell marker genes and upregulation of matrix metalloproteinases. These events in experimental models occur before aneurysm formation. Targeting smooth muscle cells to a reparative phenotype might provide a novel therapy in the treatment of aortic aneurysms.


Assuntos
Aorta/citologia , Aneurisma Aórtico/patologia , Músculo Liso Vascular/citologia , Actinas/análise , Animais , Aorta Abdominal/citologia , Aneurisma Aórtico/metabolismo , Aneurisma da Aorta Abdominal/metabolismo , Aneurisma da Aorta Abdominal/patologia , Regulação para Baixo , Expressão Gênica , Humanos , Imuno-Histoquímica , Metaloproteinase 2 da Matriz/análise , Metaloproteinase 9 da Matriz/análise , Camundongos , Camundongos Endogâmicos C57BL , Músculo Liso Vascular/química , Elastase Pancreática/farmacologia , Fenótipo , Reação em Cadeia da Polimerase , Regulação para Cima
20.
J Neurosci ; 29(46): 14451-62, 2009 Nov 18.
Artigo em Inglês | MEDLINE | ID: mdl-19923279

RESUMO

Amyloid precursor protein (APP) is a ubiquitously expressed type 1 integral membrane protein. It has the ability to bind numerous extracellular matrix components and propagate signaling responses via its cytoplasmic phospho-tyrosine, (682)YENPTY(687), binding motif. We recently demonstrated increased protein levels of APP, phosphorylated APP (Tyr682), and beta-amyloid (Abeta) in brain vasculature of atherosclerotic and Alzheimer's disease (AD) tissue colocalizing primarily within the endothelial layer. This study demonstrates similar APP changes in peripheral vasculature from human and mouse apoE(-/-) aorta, suggesting that APP-related changes are not restricted to brain vasculature. Therefore, primary mouse aortic endothelial cells and human umbilical vein endothelial cells were used as a model system to examine the function of APP in endothelial cells. APP multimerization with an anti-N-terminal APP antibody, 22C11, to simulate ligand binding stimulated an Src kinase family-dependent increase in protein phospho-tyrosine levels, APP phosphorylation, and Abeta secretion. Furthermore, APP multimerization stimulated increased protein levels of the proinflammatory proteins, cyclooxygenase-2 and vascular cell adhesion molecule-1 also in an Src kinase family-dependent manner. Endothelial APP was also involved in mediating monocytic cell adhesion. Collectively, these data demonstrate that endothelial APP regulates immune cell adhesion and stimulates a tyrosine kinase-dependent response driving acquisition of a reactive endothelial phenotype. These APP-mediated events may serve as therapeutic targets for intervention in progressive vascular changes common to cerebrovascular disease and AD.


Assuntos
Precursor de Proteína beta-Amiloide/fisiologia , Células Endoteliais/enzimologia , Proteínas Tirosina Quinases/metabolismo , Animais , Aorta Abdominal/citologia , Aorta Abdominal/enzimologia , Aorta Abdominal/imunologia , Linhagem Celular Tumoral , Células Cultivadas , Células Endoteliais/citologia , Células Endoteliais/imunologia , Ativação Enzimática/imunologia , Humanos , Imunofenotipagem , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA