Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 158
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Arterioscler Thromb Vasc Biol ; 40(7): e214-e226, 2020 07.
Artigo em Inglês | MEDLINE | ID: mdl-32493171

RESUMO

OBJECTIVE: Mitochondria consistently change their morphology in a process regulated by proteins, including Drp1 (dynamin-related protein 1), a protein promoting mitochondrial fission. Drp1 is involved in the mechanisms underlying various cardiovascular diseases, such as myocardial ischemia/reperfusion injury, heart failure, and pulmonary arterial hypertension. However, its role in macrophages, which promote various vascular diseases, is poorly understood. We therefore tested our hypothesis that macrophage Drp1 promotes vascular remodeling after injury. METHOD AND RESULTS: To explore the selective role of macrophage Drp1, we created macrophage-selective Drp1-deficient mice and performed femoral arterial wire injury. In these mice, intimal thickening and negative remodeling were attenuated at 4 weeks after injury when compared with control mice. Deletion of macrophage Drp1 also attenuated the macrophage accumulation and cell proliferation in the injured arteries. Gain- and loss-of-function experiments using cultured macrophages indicated that Drp1 induces the expression of molecules associated with inflammatory macrophages. Morphologically, mitochondrial fission was induced in inflammatory macrophages, whereas mitochondrial fusion was induced in less inflammatory/reparative macrophages. Pharmacological inhibition or knockdown of Drp1 decreased the mitochondrial reactive oxygen species and chemotactic activity in cultured macrophages. Co-culture experiments of macrophages with vascular smooth muscle cells indicated that deletion of macrophage Drp1 suppresses growth and migration of vascular smooth muscle cells induced by macrophage-derived soluble factors. CONCLUSIONS: Macrophage Drp1 accelerates intimal thickening after vascular injury by promoting macrophage-mediated inflammation. Macrophage Drp1 may be a potential therapeutic target of vascular diseases.


Assuntos
Dinaminas/metabolismo , Artéria Femoral/metabolismo , Macrófagos Peritoneais/metabolismo , Mitocôndrias/metabolismo , Neointima , Remodelação Vascular , Lesões do Sistema Vascular/metabolismo , Animais , Proliferação de Células , Quimiotaxia , Técnicas de Cocultura , Modelos Animais de Doenças , Dinaminas/deficiência , Dinaminas/genética , Artéria Femoral/lesões , Artéria Femoral/patologia , Artéria Femoral/fisiopatologia , Ativação de Macrófagos , Macrófagos Peritoneais/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Mitocôndrias/patologia , Dinâmica Mitocondrial , Músculo Liso Vascular/metabolismo , Músculo Liso Vascular/patologia , Músculo Liso Vascular/fisiopatologia , Miócitos de Músculo Liso/metabolismo , Miócitos de Músculo Liso/patologia , Células RAW 264.7 , Espécies Reativas de Oxigênio/metabolismo , Transdução de Sinais , Fatores de Tempo , Lesões do Sistema Vascular/genética , Lesões do Sistema Vascular/patologia , Lesões do Sistema Vascular/fisiopatologia
2.
Arterioscler Thromb Vasc Biol ; 38(4): 757-771, 2018 04.
Artigo em Inglês | MEDLINE | ID: mdl-29449331

RESUMO

OBJECTIVE: Plaque erosion is increasing its importance as one of the mechanisms of acute coronary syndromes in this statin era. However, the clinical efficacy of currently used lipid-lowering agents in the prevention of thrombotic complications associated with plaque erosion has not been clarified. Therefore, we examined the therapeutic effects of ezetimibe or rosuvastatin monotherapy on spontaneous atherothrombotic occlusion. APPROACH AND RESULTS: Femoral arteries of Japanese white rabbits, fed a high-cholesterol diet, were injured by balloon catheter, and then angiotensin II was continuously administrated. In 94% of these arteries, spontaneous thrombotic occlusions were observed after 5 weeks (median) of balloon injury. Histochemical analyses indicated that the injured arteries had similar pathological features to human plaque erosions; (1) spontaneous thrombotic occlusion, (2) lack of endothelial cells, and (3) tissue factor expression in vascular smooth muscle cells. Ezetimibe (1.0 mg/kg per day), but not rosuvastatin (0.6 mg/kg per day), significantly decreased thrombotic occlusion of arteries accompanied with accelerated re-endothelialization and the decreases of serum oxysterols despite the comparable on-treatment serum cholesterol levels. The 7-ketocholesterol inhibited the migration of human umbilical vein endothelial cells. Both 7-ketocholesterol and 27-hydroxycholesterol increased tissue factor expression in cultured rat vascular smooth muscle cells. Tissue factor expression was also induced by serum from vehicle- or rosuvastatin-treated rabbits, but the induction was attenuated with serum from ezetimibe-treated rabbits. CONCLUSIONS: We have established a novel rabbit model of spontaneous atherothromobotic occlusion without plaque rupture that is feasible to test the therapeutic effects of various pharmacotherapies. Ezetimibe may decrease atherothrombotic complications after superficial plaque erosion by reducing serum oxysterols.


Assuntos
Anticolesterolemiantes/farmacologia , Arteriopatias Oclusivas/prevenção & controle , Aterosclerose/tratamento farmacológico , Ezetimiba/farmacologia , Artéria Femoral/efeitos dos fármacos , Oxisteróis/sangue , Placa Aterosclerótica , Trombose/prevenção & controle , Lesões do Sistema Vascular/tratamento farmacológico , Angiotensina II , Animais , Arteriopatias Oclusivas/sangue , Arteriopatias Oclusivas/patologia , Aterosclerose/sangue , Aterosclerose/patologia , Biomarcadores/sangue , Células Cultivadas , Colesterol na Dieta , Dieta Hiperlipídica , Modelos Animais de Doenças , Regulação para Baixo , Artéria Femoral/metabolismo , Artéria Femoral/patologia , Humanos , Inibidores de Hidroximetilglutaril-CoA Redutases/farmacologia , Masculino , Coelhos , Ratos , Rosuvastatina Cálcica/farmacologia , Transdução de Sinais/efeitos dos fármacos , Trombose/sangue , Trombose/patologia , Lesões do Sistema Vascular/sangue , Lesões do Sistema Vascular/patologia
3.
J Vasc Surg ; 68(2): 576-586.e1, 2018 08.
Artigo em Inglês | MEDLINE | ID: mdl-28974341

RESUMO

OBJECTIVES: Budding uninhibited by benzimidazole-related 1 (BubR1), a cell cycle-related protein, is an essential component of the spindle checkpoint that regulates cell division. Mice in which BubR1 expression is reduced to 10% of the normal level display the phenotypic features of progeria. However, the role of BubR1 in vascular diseases and angiogenesis remains unknown. To investigate the influence of BubR1 on angiogenesis, we generated a low-null-BubR1-expressing (BubR1L/-) mouse strain with reduced BubR1 expression as low as 15% of the normal level without any abnormalities in appearance. METHODS: To elucidate the role of BubR1 in angiogenesis, we used a hind limb ischemia model induced in BubR1L/- mice and age-matched wild-type (WT) littermates. To evaluate the pathologic influence of BubR1 on angiogenesis, we measured the blood flow before and after hind limb ischemia surgery, and the expression of typical angiogenic factors in vivo and in vitro. RESULTS: In WT mice, blood flow in the ischemic left limb gradually recovered to approximately 80%, 14 days after surgery. Conversely, in the BubR1L/- group, blood flow in the left ischemic limb recovered to at most 30% (14 days after surgery, P < .01; immediately after the operation, and 5 and 9 days after surgery, P < .05). In adductor and calf muscles from BubR1L/- mice, regenerated muscle bundles, granulation tissue, and inflammatory cell invasion were more evident than in calf muscles from WT mice at 14 days after surgery. All WT mice at 14 days after surgery had complete limb salvage, but loss of limbs was observed in approximately 70% of BubR1L/- mice (P < .05). The vascular endothelial growth factor protein increase in ischemic hind limb muscles was lower in BubR1L/- mice compared with WT mice (P < .05), and vascular endothelial growth factor levels in human aortic smooth muscle cells treated with BubR1 knockdown siRNA were lower compared with scramble siRNA under hypoxic conditions (P < .01). HIF1α protein levels in the muscles after hind limb ischemia surgery were also significantly lower in BubR1L/- mice compared with WT mice (P < .05). CONCLUSIONS: BubR1 insufficiency impairs angiogenesis and results in limb loss in ischemic hind limbs. BubR1 may be a crucial angiogenic factor and might be beneficial for the treatment of limb ischemia.


Assuntos
Envelhecimento/metabolismo , Proteínas de Ciclo Celular/deficiência , Isquemia/metabolismo , Músculo Esquelético/irrigação sanguínea , Músculo Esquelético/metabolismo , Neovascularização Fisiológica , Proteínas Serina-Treonina Quinases/deficiência , Fatores Etários , Envelhecimento/genética , Animais , Proteínas de Ciclo Celular/genética , Células Cultivadas , Estado Terminal , Modelos Animais de Doenças , Membro Posterior , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Isquemia/genética , Isquemia/fisiopatologia , Masculino , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Knockout , Músculo Liso Vascular/metabolismo , Miócitos de Músculo Liso/metabolismo , Neovascularização Fisiológica/genética , Proteínas Serina-Treonina Quinases/genética , Proteínas Serina-Treonina Quinases/metabolismo , Interferência de RNA , Fluxo Sanguíneo Regional , Transdução de Sinais , Fatores de Tempo , Transfecção , Fator A de Crescimento do Endotélio Vascular/metabolismo
4.
Arterioscler Thromb Vasc Biol ; 37(2): 350-358, 2017 02.
Artigo em Inglês | MEDLINE | ID: mdl-27932353

RESUMO

OBJECTIVES: We sought to investigate whether treatment with ezetimibe in combination with statins improves coronary endothelial function in target vessels in coronary artery disease patients after coronary stenting. APPROACH AND RESULTS: We conducted a multicenter, prospective, randomized, open-label, blinded-end point trial among 11 cardiovascular treatment centers. From 2011 to 2013, 260 coronary artery disease patients who underwent coronary stenting were randomly allocated to 2 arms (statin monotherapy, S versus ezetimibe [10 mg/d]+statin combinational therapy, E+S). We defined target vessel dysfunction as the primary composite outcome, which comprised target vessel failure during treatment and at the 6- to 8-month follow-up coronary angiography and coronary endothelial dysfunction determined via intracoronary acetylcholine testing performed in cases without target vessel failure at the follow-up coronary angiography. Coadministration of ezetimibe with statins further lowered low-density lipoprotein cholesterol levels (83±23 mg/dL in S versus 67±23 mg/dL in E+S; P<0.0001), with significant decreases in oxidized low-density lipoprotein and oxysterol levels. Among patients without target vessel failure, 46 out of 89 patients (52%) in the S arm and 34 out of 96 patients (35%) in the E+S arm were found to have coronary endothelial dysfunction (P=0.0256), and the incidence of target vessel dysfunction at follow-up was significantly decreased in the E+S arm (69/112 (62%) in S versus 47/109 (43%) in E+S; P=0.0059). A post hoc analysis of post-treatment low-density lipoprotein cholesterol-matched subgroups revealed that the incidence of both target vessel dysfunction and coronary endothelial dysfunction significantly decreased in the E+S arm, with significant reductions in oxysterol levels. CONCLUSIONS: The CuVIC trial (Effect of Cholesterol Absorption Inhibitor Usage on Target Vessel Dysfunction after Coronary Stenting) has shown that ezetimibe with statins, compared with statin monotherapy, improves functional prognoses, ameliorating endothelial dysfunction in stented coronary arteries, and was associated with larger decreases in oxysterol levels.


Assuntos
Anticolesterolemiantes/uso terapêutico , Doença da Artéria Coronariana/terapia , Vasos Coronários/efeitos dos fármacos , Endotélio Vascular/efeitos dos fármacos , Ezetimiba/uso terapêutico , Inibidores de Hidroximetilglutaril-CoA Redutases/uso terapêutico , Intervenção Coronária Percutânea/instrumentação , Stents , Acetilcolina/administração & dosagem , Idoso , Anticolesterolemiantes/efeitos adversos , Biomarcadores/sangue , LDL-Colesterol/sangue , Angiografia Coronária , Doença da Artéria Coronariana/sangue , Doença da Artéria Coronariana/diagnóstico por imagem , Doença da Artéria Coronariana/fisiopatologia , Vasos Coronários/diagnóstico por imagem , Vasos Coronários/fisiopatologia , Combinação de Medicamentos , Endotélio Vascular/fisiopatologia , Ezetimiba/efeitos adversos , Feminino , Humanos , Inibidores de Hidroximetilglutaril-CoA Redutases/efeitos adversos , Japão , Lipoproteínas LDL/sangue , Masculino , Pessoa de Meia-Idade , Oxisteróis/sangue , Intervenção Coronária Percutânea/efeitos adversos , Estudos Prospectivos , Fatores de Tempo , Resultado do Tratamento
5.
Int Heart J ; 59(6): 1432-1444, 2018 Nov 28.
Artigo em Inglês | MEDLINE | ID: mdl-30369578

RESUMO

Statins are known to improve pulmonary arterial hypertension (PAH) by their anti-inflammatory and anti-proliferative effects in animal models. However, recent clinical studies have reported that clinically approved statin doses failed to improve clinical outcomes in patients with PAH. We therefore hypothesized that nanoparticle (NP) -mediated targeting of pitavastatin could attenuate the progression of established PAH.We induced PAH by subcutaneously injecting monocrotaline (MCT) in Sprague-Dawley rats. On day 14 after the MCT injection, animals that displayed established PAH on echocardiography were included. On day 17, they were randomly assigned to the following 5 groups: daily intravenous administration of (1) vehicle, (2) fluorescein-isothiocyanate-NP, (3) pitavastatin, (4) pitavastatin-NP, or (5) oral sildenafil. Intravenous NP was selectively delivered to small pulmonary arteries and circulating CD11b-positive leukocytes. On day 21, pitavastatin-NP attenuated the progression of PAH at lower doses than pitavastatin alone. This was associated with the inhibition of monocyte-mediated inflammation, proliferation, and remodeling of the pulmonary arteries. Interestingly, sildenafil attenuated the development of PAH, but had no effects on inflammation or remodeling of the pulmonary arteries. In separate experiments, only treatment with pitavastatin-NP reduced the mortality rate at day 35.NP-mediated targeting of pitavastatin to small pulmonary arteries and leukocytes attenuated the progression of established MCT-induced PAH and improved survival. Therapeutically, pitavastatin-NP was associated with anti-inflammatory and anti-proliferative effects on small pulmonary arteries, which was completely distinct from the vasodilatory effect of sildenafil. Pitavastatin-NP can be a novel therapeutic modality for PAH.


Assuntos
Sistemas de Liberação de Medicamentos/métodos , Inibidores de Hidroximetilglutaril-CoA Redutases/administração & dosagem , Hipertensão Pulmonar/tratamento farmacológico , Nanopartículas/administração & dosagem , Quinolinas/administração & dosagem , Administração Intravenosa , Animais , Progressão da Doença , Inibidores de Hidroximetilglutaril-CoA Redutases/uso terapêutico , Hipertensão Pulmonar/induzido quimicamente , Leucócitos , Masculino , Monocrotalina , Artéria Pulmonar , Quinolinas/uso terapêutico , Distribuição Aleatória , Ratos , Ratos Sprague-Dawley , Resultado do Tratamento
6.
Int Heart J ; 59(5): 1015-1025, 2018 Sep 26.
Artigo em Inglês | MEDLINE | ID: mdl-30158384

RESUMO

Pulmonary hypertension (PH) is a disease with poor prognosis, caused by the obstruction/stenosis of small pulmonary arteries. Statin is known to have vasodilating and anti-inflammatory property and is considered to be a candidate of therapeutic agents for the treatment of PH, but its efficacy has not been verified in clinical trials. We have formulated pitavastatin incorporating nanoparticles composed of poly (lactic-co-glycolic acid) (NK-104-NP) to improve drug delivery to the pulmonary arteries and evaluated their safety and pharmacokinetics in healthy volunteers. To accomplish this purpose, phase I clinical trials were conducted. In the single intravenous administration regimen, 40 healthy subjects were enrolled and PK (pharmacokinetic) parameters in 4 groups (1, 2, 4, and 8 mg as pitavastatin calcium) were as follows: 1.00 hour after the administration, the plasma concentration of pitavastatin reached Cmax (the maximum drug concentration) in all groups. Cmax, AUC0-t (area under the curve from time 0 to the last measurable concentration) and AUC0-∞ (area under the curve from time 0 extrapolated to infinite time) were increased in a dose-dependent manner. Population pharmacokinetic analysis based on these results indicated no accumulation of pitavastatin after repeated administration of NK-104-NP for 7 days. In this 7-day administration trial, the mean Cmax and AUC0-∞ of pitavastatin were not significantly different between days 1 and 7, suggesting that pitavastatin is unlikely to accumulate after repeated administration. In these trials, three adverse events (AEs) were reported, but they were resolved without any complications and judged to have no causal relationships with NK-104-NP. These results indicate that the innovative nanotechnology-based medicine NK-104-NP exhibited dose-dependent pharmacokinetics and was well tolerated with no significant AEs in healthy volunteers.


Assuntos
Inibidores de Hidroximetilglutaril-CoA Redutases/farmacocinética , Hipertensão Pulmonar/tratamento farmacológico , Pulmão/irrigação sanguínea , Quinolinas/farmacocinética , Administração Intravenosa , Adulto , Sistemas de Liberação de Medicamentos , Voluntários Saudáveis , Humanos , Inibidores de Hidroximetilglutaril-CoA Redutases/administração & dosagem , Inibidores de Hidroximetilglutaril-CoA Redutases/uso terapêutico , Hipertensão Pulmonar/mortalidade , Hipertensão Pulmonar/patologia , Japão/epidemiologia , Pulmão/efeitos dos fármacos , Pulmão/patologia , Masculino , Nanotecnologia/métodos , Artéria Pulmonar/efeitos dos fármacos , Quinolinas/administração & dosagem , Quinolinas/sangue , Quinolinas/uso terapêutico
7.
Circulation ; 133(13): 1249-63, 2016 Mar 29.
Artigo em Inglês | MEDLINE | ID: mdl-26915633

RESUMO

BACKGROUND: Mitochondrial autophagy is an important mediator of mitochondrial quality control in cardiomyocytes. The occurrence of mitochondrial autophagy and its significance during cardiac hypertrophy are not well understood. METHODS AND RESULTS: Mice were subjected to transverse aortic constriction (TAC) and observed at multiple time points up to 30 days. Cardiac hypertrophy developed after 5 days, the ejection fraction was reduced after 14 days, and heart failure was observed 30 days after TAC. General autophagy was upregulated between 1 and 12 hours after TAC but was downregulated below physiological levels 5 days after TAC. Mitochondrial autophagy, evaluated by electron microscopy, mitochondrial content, and Keima with mitochondrial localization signal, was transiently activated at ≈3 to 7 days post-TAC, coinciding with mitochondrial translocation of Drp1. However, it was downregulated thereafter, followed by mitochondrial dysfunction. Haploinsufficiency of Drp1 abolished mitochondrial autophagy and exacerbated the development of both mitochondrial dysfunction and heart failure after TAC. Injection of Tat-Beclin 1, a potent inducer of autophagy, but not control peptide, on day 7 after TAC, partially rescued mitochondrial autophagy and attenuated mitochondrial dysfunction and heart failure induced by overload. Haploinsufficiency of either drp1 or beclin 1 prevented the rescue by Tat-Beclin 1, suggesting that its effect is mediated in part through autophagy, including mitochondrial autophagy. CONCLUSIONS: Mitochondrial autophagy is transiently activated and then downregulated in the mouse heart in response to pressure overload. Downregulation of mitochondrial autophagy plays an important role in mediating the development of mitochondrial dysfunction and heart failure, whereas restoration of mitochondrial autophagy attenuates dysfunction in the heart during pressure overload.


Assuntos
Autofagia/fisiologia , Dinaminas/metabolismo , Insuficiência Cardíaca/metabolismo , Insuficiência Cardíaca/prevenção & controle , Mitocôndrias/metabolismo , Sequência de Aminoácidos , Animais , Dinaminas/genética , Insuficiência Cardíaca/patologia , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Mitocôndrias/patologia , Dados de Sequência Molecular , Pressão
8.
Circ Res ; 116(2): 264-78, 2015 Jan 16.
Artigo em Inglês | MEDLINE | ID: mdl-25332205

RESUMO

RATIONALE: Both fusion and fission contribute to mitochondrial quality control. How unopposed fusion affects survival of cardiomyocytes and left ventricular function in the heart is poorly understood. OBJECTIVE: We investigated the role of dynamin-related protein 1 (Drp1), a GTPase that mediates mitochondrial fission, in mediating mitochondrial autophagy, ventricular function, and stress resistance in the heart. METHODS AND RESULTS: Drp1 downregulation induced mitochondrial elongation, accumulation of damaged mitochondria, and increased apoptosis in cardiomyocytes at baseline. Drp1 downregulation also suppressed autophagosome formation and autophagic flux at baseline and in response to glucose deprivation in cardiomyocytes. The lack of lysosomal translocation of mitochondrially targeted Keima indicates that Drp1 downregulation suppressed mitochondrial autophagy. Mitochondrial elongation and accumulation of damaged mitochondria were also observed in tamoxifen-inducible cardiac-specific Drp1 knockout mice. After Drp1 downregulation, cardiac-specific Drp1 knockout mice developed left ventricular dysfunction, preceded by mitochondrial dysfunction, and died within 13 weeks. Autophagic flux is significantly suppressed in cardiac-specific Drp1 knockout mice. Although left ventricular function in cardiac-specific Drp1 heterozygous knockout mice was normal at 12 weeks of age, left ventricular function decreased more severely after 48 hours of fasting, and the infarct size/area at risk after ischemia/reperfusion was significantly greater in cardiac-specific Drp1 heterozygous knockout than in control mice. CONCLUSIONS: Disruption of Drp1 induces mitochondrial elongation, inhibits mitochondrial autophagy, and causes mitochondrial dysfunction, thereby promoting cardiac dysfunction and increased susceptibility to ischemia/reperfusion.


Assuntos
Autofagia/fisiologia , Dinaminas/fisiologia , Metabolismo Energético/fisiologia , Mitocôndrias Cardíacas/metabolismo , Estresse Oxidativo/fisiologia , Animais , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Miócitos Cardíacos/fisiologia , Ratos , Ratos Wistar
9.
Arterioscler Thromb Vasc Biol ; 36(3): 491-500, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26821947

RESUMO

OBJECTIVE: Inflammatory monocytes/macrophages produce various proteinases, including matrix metalloproteinases, and degradation of the extracellular matrix by these activated proteinases weakens the mechanical strength of atherosclerotic plaques, which results in a rupture of the plaque. Peroxisome proliferator-activated receptor-γ induces a polarity shift of monocytes/macrophages toward less inflammatory phenotypes and has the potential to prevent atherosclerotic plaque ruptures. Therefore, we hypothesized that nanoparticle-mediated targeted delivery of the peroxisome proliferator-activated receptor-γ agonist pioglitazone into circulating monocytes could effectively inhibit plaque ruptures in a mouse model. APPROACH AND RESULTS: We prepared bioabsorbable poly(lactic-co-glycolic-acid) nanoparticles containing pioglitazone (pioglitazone-NPs). Intravenously administered poly(lactic-co-glycolic-acid) nanoparticles incorporated with fluorescein isothiocyanate were found in circulating monocytes and aortic macrophages by flow cytometric analysis. Weekly intravenous administration of pioglitazone-NPs (7 mg/kg per week) for 4 weeks decreased buried fibrous caps, a surrogate marker of plaque rupture, in the brachiocephalic arteries of ApoE(-/-) mice fed a high-fat diet and infused with angiotensin II. In contrast, administration of control-NPs or an equivalent dose of oral pioglitazone treatment produced no effects. Pioglitazone-NPs inhibited the activity of matrix metalloproteinases and cathepsins in the brachiocephalic arteries. Pioglitazone-NPs regulated inflammatory cytokine expression and also suppressed the expression of extracellular matrix metalloproteinase inducer in bone marrow-derived macrophages. CONCLUSIONS: Nanoparticle-mediated delivery of pioglitazone inhibited macrophage activation and atherosclerotic plaque ruptures in hyperlipidemic ApoE(-/-) mice. These results demonstrate a promising strategy with a favorable safety profile to prevent atherosclerotic plaque ruptures.


Assuntos
Aterosclerose/tratamento farmacológico , Fármacos Cardiovasculares/farmacologia , Diferenciação Celular/efeitos dos fármacos , Portadores de Fármacos , Ácido Láctico/química , Macrófagos Peritoneais/efeitos dos fármacos , Monócitos/efeitos dos fármacos , Nanopartículas , Placa Aterosclerótica , Ácido Poliglicólico/química , Tiazolidinedionas/farmacologia , Administração Intravenosa , Angiotensina II , Animais , Apolipoproteínas E/deficiência , Apolipoproteínas E/genética , Aterosclerose/induzido quimicamente , Aterosclerose/genética , Aterosclerose/metabolismo , Aterosclerose/patologia , Tronco Braquiocefálico/efeitos dos fármacos , Tronco Braquiocefálico/metabolismo , Tronco Braquiocefálico/patologia , Fármacos Cardiovasculares/administração & dosagem , Fármacos Cardiovasculares/química , Catepsinas/metabolismo , Células Cultivadas , Química Farmacêutica , Citocinas/metabolismo , Dieta Hiperlipídica , Modelos Animais de Doenças , Progressão da Doença , Mediadores da Inflamação/metabolismo , Macrófagos Peritoneais/metabolismo , Macrófagos Peritoneais/patologia , Masculino , Metaloproteinases da Matriz/metabolismo , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Knockout , Monócitos/metabolismo , Monócitos/patologia , Fenótipo , Pioglitazona , Copolímero de Ácido Poliláctico e Ácido Poliglicólico , Ruptura Espontânea , Tiazolidinedionas/administração & dosagem , Tiazolidinedionas/química
10.
Int Heart J ; 58(4): 615-623, 2017 Aug 03.
Artigo em Inglês | MEDLINE | ID: mdl-28701679

RESUMO

Left ventricular (LV) remodeling after myocardial infarction (MI) causes heart failure. Although medical therapies including angiotensin converting enzyme inhibitors show inhibitory effects on post-infarct LV remodeling, the prognosis of patients with post-infarct heart failure is still poor. Accumulating evidence suggests that an inflammatory response is implicated in the process of post-infarct LV remodeling. Therefore, we hypothesized that anti-inflammatory therapy by nanoparticle-mediated monocyte/macrophage-targeting delivery of pitavastatin may protect the heart from post-infarct LV remodeling.Male C57BL/6 mice were subjected to permanent coronary ligation and pitavastatin-incorporating nanoparticles (Pitavastatin-NPs) were intravenously injected for 3 to 5 consecutive days. Pitavastatin-NPs were delivered to CD11b+ monocytes/macrophages, but not to cardiomyocytes. Treatment with Pitavastatin-NPs after establishment of MI attenuated post-infarct LV remodeling accompanied by a reduction of monocytes/macrophages in the heart, whereas pitavastatin solution treatment did not. Pitavastatin-NPs inhibited mobilization of monocytes from the spleen after MI. In mice after splenectomy, Pitavastatin-NPs still decreased the number of monocytes/macrophages in the infarcted heart and inhibited post-infarct LV remodeling.Nanoparticle-mediated delivery of pitavastatin to monocytes/macrophages may be a novel therapeutic strategy to protect the heart from post-infarct LV remodeling. Inhibition of monocyte mobilization from the bone marrow is one of the major mechanisms by which Pitavastatin-NPs attenuated post-infarct LV remodeling.


Assuntos
Sistemas de Liberação de Medicamentos , Insuficiência Cardíaca/prevenção & controle , Inflamação/tratamento farmacológico , Infarto do Miocárdio/fisiopatologia , Nanopartículas/administração & dosagem , Quinolinas/administração & dosagem , Remodelação Ventricular/efeitos dos fármacos , Animais , Modelos Animais de Doenças , Insuficiência Cardíaca/etiologia , Insuficiência Cardíaca/fisiopatologia , Inibidores de Hidroximetilglutaril-CoA Redutases/administração & dosagem , Injeções Intravenosas , Macrófagos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Monócitos , Infarto do Miocárdio/complicações
11.
J Cardiovasc Pharmacol ; 67(4): 290-8, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26745002

RESUMO

Nanoparticles (NPs) have been used as novel drug delivery systems. Drug-incorporated NPs for local delivery might optimize the efficacy and minimize the side effects of drugs. Intravenous prostacyclin improves long-term survival in patients with pulmonary arterial hypertension (PAH), but it causes serious side effects such as catheter-related infections. We investigated the efficacy and safety of intratracheal administration of a prostacyclin analogue, beraprost (BPS), incorporated NPs in Sugen-hypoxia-normoxia and monocrotaline rat models of PAH and in human PAH pulmonary arterial smooth muscle cells (PASMCs). After a single administration, BPS NPs significantly decreased right ventricular pressure, right ventricular hypertrophy, and pulmonary artery muscularization in the 2 rat models. BPS NPs significantly improved the survival rate in the monocrotaline rat model. No infiltration of inflammatory cells, hemorrhage, or fibrosis was found in the liver, kidney, spleen, and heart after the administration of BPS NPs. No liver or kidney dysfunction was found in the blood examinations. BPS and BPS NPs significantly inhibited the proliferation of human PAH PASMCs after 24 hours of treatment. BPS NPs significantly continued to inhibit the proliferation of human PAH PASMCs at 24 hours after the removal of BPS NPs. BPS NPs significantly induced apoptosis in PAH PASMCs compared to that in non-PAH PASMCs. Intratracheal administration of BPS NPs ameliorates pulmonary hypertension in PAH rat models by a sustained antiproliferative effect and a proapoptotic effect on PAH PASMCs.


Assuntos
Epoprostenol/análogos & derivados , Hipertensão Pulmonar/prevenção & controle , Nanopartículas , Adolescente , Adulto , Animais , Hipóxia Celular , Proliferação de Células/efeitos dos fármacos , Criança , Modelos Animais de Doenças , Sistemas de Liberação de Medicamentos , Epoprostenol/administração & dosagem , Epoprostenol/farmacologia , Feminino , Humanos , Hipertrofia Ventricular Direita/tratamento farmacológico , Hipertrofia Ventricular Direita/patologia , Masculino , Monocrotalina/toxicidade , Miócitos de Músculo Liso/efeitos dos fármacos , Miócitos de Músculo Liso/metabolismo , Artéria Pulmonar/efeitos dos fármacos , Artéria Pulmonar/metabolismo , Ratos , Ratos Sprague-Dawley , Fatores de Tempo , Adulto Jovem
12.
Circulation ; 129(8): 896-906, 2014 Feb 25.
Artigo em Inglês | MEDLINE | ID: mdl-24305567

RESUMO

BACKGROUND: Preventing atherosclerotic plaque destabilization and rupture is the most reasonable therapeutic strategy for acute myocardial infarction. Therefore, we tested the hypotheses that (1) inflammatory monocytes play a causative role in plaque destabilization and rupture and (2) the nanoparticle-mediated delivery of pitavastatin into circulating inflammatory monocytes inhibits plaque destabilization and rupture. METHODS AND RESULTS: We used a model of plaque destabilization and rupture in the brachiocephalic arteries of apolipoprotein E-deficient (ApoE(-/-)) mice fed a high-fat diet and infused with angiotensin II. The adoptive transfer of CCR2(+/+)Ly-6C(high) inflammatory macrophages, but not CCR2(-/-) leukocytes, accelerated plaque destabilization associated with increased serum monocyte chemoattractant protein-1 (MCP-1), monocyte-colony stimulating factor, and matrix metalloproteinase-9. We prepared poly(lactic-co-glycolic) acid nanoparticles that were incorporated by Ly-6G(-)CD11b(+) monocytes and delivered into atherosclerotic plaques after intravenous administration. Intravenous treatment with pitavastatin-incorporated nanoparticles, but not with control nanoparticles or pitavastatin alone, inhibited plaque destabilization and rupture associated with decreased monocyte infiltration and gelatinase activity in the plaque. Pitavastatin-incorporated nanoparticles inhibited MCP-1-induced monocyte chemotaxis and the secretion of MCP-1 and matrix metalloproteinase-9 from cultured macrophages. Furthermore, the nanoparticle-mediated anti-MCP-1 gene therapy reduced the incidence of plaque destabilization and rupture. CONCLUSIONS: The recruitment of inflammatory monocytes is critical in the pathogenesis of plaque destabilization and rupture, and nanoparticle-mediated pitavastatin delivery is a promising therapeutic strategy to inhibit plaque destabilization and rupture by regulating MCP-1/CCR2-dependent monocyte recruitment in this model.


Assuntos
Sistemas de Liberação de Medicamentos/métodos , Monócitos/efeitos dos fármacos , Nanopartículas/metabolismo , Placa Aterosclerótica/tratamento farmacológico , Quinolinas/farmacocinética , Transferência Adotiva , Animais , Apolipoproteínas E/genética , Tronco Braquiocefálico/efeitos dos fármacos , Tronco Braquiocefálico/imunologia , Tronco Braquiocefálico/patologia , Movimento Celular/efeitos dos fármacos , Movimento Celular/imunologia , Quimiocina CCL2/antagonistas & inibidores , Quimiocina CCL2/metabolismo , Modelos Animais de Doenças , Terapia Genética/métodos , Inibidores de Hidroximetilglutaril-CoA Redutases/farmacocinética , Masculino , Camundongos , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Knockout , Monócitos/imunologia , Monócitos/patologia , Nanopartículas/uso terapêutico , Placa Aterosclerótica/imunologia , Placa Aterosclerótica/patologia , Receptores CCR2/genética
13.
Int Heart J ; 56(3): 354-9, 2015 May 13.
Artigo em Inglês | MEDLINE | ID: mdl-25902888

RESUMO

Platelet-derived growth factor (PDGF) is implicated in the pathogenesis of pulmonary arterial hypertension (PAH). Imatinib, a PDGF-receptor tyrosine kinase inhibitor, improved hemodynamics, but serious side effects and drug discontinuation are common when treating PAH. A drug delivery system using nanoparticles (NPs) enables the reduction of side effects while maintaining the effects of the drug. We examined the efficacy of imatinib-incorporated NPs (Ima-NPs) in a rat model and in human PAH-pulmonary arterial smooth muscle cells (PASMCs). Rats received a single intratracheal administration of PBS, FITC-NPs, or Ima-NPs immediately after monocrotaline injection. Three weeks after monocrotaline injection, intratracheal administration of Ima-NPs suppressed the development of pulmonary hypertension, small pulmonary artery remodeling, and right ventricular hypertrophy in the rat model of monocrotaline-induced PAH. We also examined the effects of imatinib and Ima-NPs on PDGF-induced proliferation of human PAH-PASMCs by (3)H-thymidine incorporation. Imatinib and Ima-NPs significantly inhibited proliferation after 24 hours of treatment. Ima-NPs significantly inhibited proliferation compared with imatinib at 24 hours after removal of these drugs. Delivery of Ima-NPs into lungs suppressed the development of MCT-induced PAH by sustained antiproliferative effects on PAS-MCs.


Assuntos
Benzamidas/administração & dosagem , Hipertensão Pulmonar/prevenção & controle , Nanopartículas , Piperazinas/administração & dosagem , Pirimidinas/administração & dosagem , Receptores do Fator de Crescimento Derivado de Plaquetas/antagonistas & inibidores , Animais , Benzamidas/uso terapêutico , Células Cultivadas , Modelos Animais de Doenças , Humanos , Hipertensão Pulmonar/induzido quimicamente , Mesilato de Imatinib , Masculino , Monocrotalina , Músculo Liso Vascular/citologia , Miócitos de Músculo Liso/efeitos dos fármacos , Piperazinas/uso terapêutico , Pirimidinas/uso terapêutico , Ratos , Ratos Sprague-Dawley
14.
J Cell Sci ; 125(Pt 10): 2407-15, 2012 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-22357958

RESUMO

Age-related macular degeneration (AMD) is the leading cause of blindness among the elderly in developed countries. Although pathogenic factors, such as oxidative stress, inflammation and genetics are thought to contribute to the development of AMD, little is known about the relationships and priorities between these factors. Here, we show that chronic photo-oxidative stress is an environmental factor involved in AMD pathogenesis. We first demonstrated that exposure to light induced phospholipid oxidation in the mouse retina, which was more prominent in aged animals. The induced oxidized phospholipids led to an increase in the expression of monocyte chemoattractant protein-1, which then resulted in macrophage accumulation, an inflammatory process. Antioxidant treatment prevented light-induced phospholipid oxidation and the subsequent increase of monocyte chemoattractant protein-1 (also known as C-C motif chemokine 2; CCL2), which are the beginnings of the light-induced changes. Subretinal application of oxidized phospholipids induced choroidal neovascularization, a characteristic feature of wet-type AMD, which was inhibited by blocking monocyte chemoattractant protein-1. These findings strongly suggest that a sequential cascade from photic stress to inflammatory processes through phospholipid oxidation has an important role in AMD pathogenesis. Finally, we succeeded in mimicking human AMD in mice with low-level, long-term photic stress, in which characteristic pathological changes, including choroidal neovascularization formation, were observed. Therefore, we propose a consecutive pathogenic pathway involving photic stress, oxidation of phospholipids and chronic inflammation, leading to angiogenesis. These findings add to the current understanding of AMD pathology and suggest protection from oxidative stress or suppression of the subsequent inflammation as new potential therapeutic targets for AMD.


Assuntos
Quimiocina CCL2/metabolismo , Degeneração Macular/metabolismo , Estresse Oxidativo/efeitos da radiação , Animais , Quimiocina CCL2/genética , Modelos Animais de Doenças , Feminino , Humanos , Luz , Macrófagos/metabolismo , Degeneração Macular/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout
15.
Am J Pathol ; 183(1): 313-25, 2013 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-23685110

RESUMO

Chitinase 1 (CHIT1) is secreted by activated macrophages. Chitinase activity is raised in atherosclerotic patient sera and is present in atherosclerotic plaque. However, the role of CHIT1 in atherosclerosis is unknown. Preliminary studies of atherosclerosis in cynomolgous monkeys revealed CHIT1 to be closely correlated with areas of macrophage infiltration. Thus, we investigated the effects of a chitinase inhibitor, allosamidin, on macrophage function in vitro and on atherosclerotic development in vivo. In RAW264.7 cells, allosamidin elevated monocyte chemoattractant protein 1 and tumor necrosis factor alpha expression, and increased activator protein 1 and nuclear factor-κB transcriptional activity. Although inducible nitric oxide synthase, IL-6, and IL-1ß expression were increased, Arg1 expression was decreased by chitinase inhibition, suggesting that suppression of CHIT1 activity polarizes macrophages into a M1 phenotype. Allosamidin decreased scavenger receptor AI, CD36, ABCA1, and ABCG1 expression which led to suppression of cholesterol uptake and apolipoprotein AI-mediated cholesterol efflux in macrophages. These effects were confirmed with CHIT1 siRNA transfection and CHIT1 plasmid transfection experiments in primary macrophages. Apolipoprotein E-deficient hyperlipidemic mice treated for 6 weeks with constant administration of allosamidin and fed an atherogenic diet showed aggravated atherosclerotic lesion formation. These data suggest that CHIT1 exerts protective effects against atherosclerosis by suppressing inflammatory responses and polarizing macrophages toward an M2 phenotype, and promoting lipid uptake and cholesterol efflux in macrophages.


Assuntos
Acetilglucosamina/análogos & derivados , Aterosclerose/enzimologia , Quitinases/antagonistas & inibidores , Inibidores Enzimáticos/efeitos adversos , Macrófagos/enzimologia , Trissacarídeos/efeitos adversos , Acetilglucosamina/efeitos adversos , Animais , Aterosclerose/induzido quimicamente , Aterosclerose/metabolismo , Aterosclerose/prevenção & controle , Biomarcadores/metabolismo , Linhagem Celular , Quitinases/metabolismo , Citocinas/metabolismo , Macrófagos/efeitos dos fármacos , Macrófagos/metabolismo , Masculino , Camundongos , Camundongos Knockout , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa
16.
Int Heart J ; 55(4): 281-6, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24942639

RESUMO

Administration of drugs and other therapeutic agents has been the central strategy of contemporary medicine for cardiovascular disease. The use of a drug delivery system (DDS) is always demanded to enhance the efficacy and safety of therapeutic agents, and improve the signal-to-noise ratio of imaging agents. Nano-scale materials modify in vivo drug kinetics, depending on (patho)physiological mechanisms such as vascular permeability and incorporation by the mononuclear phagocyte system, which constitute 'passive-targeting' properties of nano-DDS. By contrast, an 'active-targeting' strategy employs a specific targeting structure on nano-DDS, which binds to the target molecule that is specific for a certain disease process, such as tumor specific antigens and the induction of adhesion molecules. In this review, we summarize recent studies that applied nano-DDS for the diagnosis and treatment of cardiovascular disease, especially focusing on atherosclerosis and myocardial ischemia-reperfusion (IR) injury. Pathophysiological changes in atherosclerosis and myocardial IR injury are successfully targeted by nano-DDS and preclinical studies in animals showed positive effects of nano-DDS enhancing efficacy and reducing adverse effects. The development of nano-DDS in clinical medicine is keenly being awaited.


Assuntos
Fármacos Cardiovasculares/administração & dosagem , Doenças Cardiovasculares/tratamento farmacológico , Sistemas de Liberação de Medicamentos , Nanopartículas/administração & dosagem , Animais , Desenho de Equipamento , Humanos
17.
Curr Opin Lipidol ; 24(5): 419-25, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23942269

RESUMO

PURPOSE OF REVIEW: Atherosclerotic plaque destabilization and rupture is an important pathological condition that may account for approximately 70% of acute myocardial infarction cases. To analyse the mechanisms by which an atherosclerotic plaque destabilizes and ruptures and examine the effects of novel therapeutic approaches, several groups have developed mouse models of plaque rupture. RECENT FINDINGS: Findings from intracoronary imaging modalities support the role of rupture-prone 'vulnerable plaques' characterized by pathological studies as precursors of plaque rupture and acute myocardial infarction. Atherosclerotic plaques in the brachiocephalic arteries of apolipoprotein E (ApoE)-deficient mice fed a high-fat diet demonstrate several key histological features of ruptured human plaques. Angiotensin II infusion accelerates plaque destabilization and rupture, which has enabled researchers to analyse the role of pathophysiological and genetic factors that accelerate plaque destabilization and rupture and qualitatively examine the effects of experimental therapies. The plaque rupture model in the brachiocephalic arteries of ApoE-deficient mice is disputed due to dissimilarities from human plaques regarding the incidence of thrombotic occlusion and computer-simulated mechanical stress in the plaque. SUMMARY: Although no mouse model examined completely simulates the entire process of plaque rupture, the brachiocephalic artery in ApoE-deficient mice fed a high-fat diet, with or without angiotensin II infusion, is a practically feasible model for plaque rupture.


Assuntos
Aterosclerose , Modelos Animais de Doenças , Placa Aterosclerótica , Animais , Apolipoproteínas E/genética , Apolipoproteínas E/metabolismo , Aterosclerose/genética , Aterosclerose/metabolismo , Aterosclerose/patologia , Artérias Cerebrais/metabolismo , Artérias Cerebrais/patologia , Gorduras na Dieta/efeitos adversos , Gorduras na Dieta/farmacologia , Humanos , Camundongos , Placa Aterosclerótica/genética , Placa Aterosclerótica/metabolismo , Placa Aterosclerótica/patologia
18.
Arterioscler Thromb Vasc Biol ; 32(10): 2427-34, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22879581

RESUMO

OBJECTIVE: Critical limb ischemia is a severe form of peripheral artery disease (PAD) for which neither surgical revascularization nor endovascular therapy nor current medicinal therapy has sufficient therapeutic effects. Peroxisome proliferator activated receptor-γ agonists present angiogenic activity in vitro; however, systemic administration of peroxisome proliferator-activated receptor-γ agonists is hampered by its side effects, including heart failure. Here, we demonstrate that the nanoparticle (NP)-mediated delivery of the peroxisome proliferator activated receptor-γ agonist pioglitazone enhances its therapeutic efficacy on ischemia-induced neovascularization in a murine model. METHODS AND RESULTS: In a nondiabetic murine model of hindlimb ischemia, a single intramuscular injection of pioglitazone-incorporated NP (1 µg/kg) into ischemic muscles significantly improved the blood flow recovery in the ischemic limbs, significantly increasing the number of CD31-positive capillaries and α-smooth muscle actin-positive arterioles. The therapeutic effects of pioglitazone-incorporated NP were diminished by the peroxisome proliferator activated receptor-γ antagonist GW9662 and were not observed in endothelial NO synthase-deficient mice. Pioglitazone-incorporated NP induced endothelial NO synthase phosphorylation, as demonstrated by Western blot analysis, as well as expression of multiple angiogenic growth factors in vivo, including vascular endothelial growth factor-A, vascular endothelial growth factor-B, and fibroblast growth factor-1, as demonstrated by real-time polymerase chain reaction. Intramuscular injection of pioglitazone (1 µg/kg) was ineffective, and oral administration necessitated a >500 µg/kg per day dose to produce therapeutic effects equivalent to those of pioglitazone-incorporated NP. CONCLUSIONS: NP-mediated drug delivery is a novel modality that may enhance the effectiveness of therapeutic neovascularization, surpassing the effectiveness of current treatments for peripheral artery disease with critical limb ischemia.


Assuntos
Membro Posterior/irrigação sanguínea , Isquemia/tratamento farmacológico , Nanopartículas , Neovascularização Fisiológica/efeitos dos fármacos , PPAR gama/agonistas , Tiazolidinedionas/farmacologia , Tiazolidinedionas/uso terapêutico , Anilidas/farmacologia , Animais , Fator 1 de Crescimento de Fibroblastos/metabolismo , Humanos , Injeções Intramusculares , Isquemia/fisiopatologia , Ácido Láctico , Camundongos , Camundongos Knockout , Modelos Animais , Neovascularização Fisiológica/fisiologia , Óxido Nítrico Sintase Tipo III/deficiência , Óxido Nítrico Sintase Tipo III/genética , Óxido Nítrico Sintase Tipo III/metabolismo , Fosforilação , Pioglitazona , Ácido Poliglicólico , Copolímero de Ácido Poliláctico e Ácido Poliglicólico , Fluxo Sanguíneo Regional/efeitos dos fármacos , Fluxo Sanguíneo Regional/fisiologia , Tiazolidinedionas/administração & dosagem , Fator A de Crescimento do Endotélio Vascular/metabolismo , Fator B de Crescimento do Endotélio Vascular/metabolismo
20.
Am J Pathol ; 179(3): 1088-94, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21741938

RESUMO

Acute respiratory distress syndrome (ARDS) is a crippling disease with no effective therapy characterized by progressive dyspnea. Mesenchymal stem cells (MSCs) have emerged as a new therapeutic modality for ARDS because MSCs can attenuate inflammation and repair the damaged tissue by differentiating into several cell types. Macrophages participate in the development of ARDS; however, MSCs only weakly modulate macrophage function. The chemokine CCL2 is a potent inducer of macrophage recruitment and activation, and its expression is elevated in patients with ARDS. We established MSCs that are stably transduced by a lentiviral vector expressing 7ND, a dominant-negative inhibitor of CCL2, to enhance the therapeutic function of MSCs. 7ND-MSCs retained the innate properties of MSCs and produced a large amount of 7ND. Many 7ND-MSCs were detected in bleomycin-treated lungs (immunostaining 24 hours after injection), suggesting that MSCs could work as a drug delivery tool. Mice treated with 7ND-MSCs showed significantly milder weight loss, lung injury, collagen content, accumulation of inflammatory cells and inflammatory mediators that were induced by bleomycin, and subsequent survival benefit. No evidence of 7ND-MSC-induced toxicity was observed during or after treatment. Thus, inhibiting the effects of macrophages may greatly enhance the ability of MSCs to effect lung repair in ARDS.


Assuntos
Quimiocina CCL2/antagonistas & inibidores , Lesão Pulmonar/prevenção & controle , Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais/fisiologia , Animais , Antibióticos Antineoplásicos/toxicidade , Bleomicina/toxicidade , Quimiocina CCL2/genética , Terapia Genética/métodos , Vetores Genéticos , Humanos , Lentivirus , Lesão Pulmonar/induzido quimicamente , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Síndrome do Desconforto Respiratório/terapia , Transdução Genética/métodos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA