Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Cardiovasc Res ; 99(4): 716-23, 2013 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-23756254

RESUMO

AIMS: The role of vascular endothelial growth factor (VEGF-A) in atherogenesis has remained controversial. We addressed this by comparing the effects of adenoviral VEGF-A gene transfer on atherosclerosis and lipoproteins in ApoE(-/-), LDLR(-/-), LDLR(-/-)ApoE(-/-), and LDLR(-/-)ApoB(100/100) mice. METHODS AND RESULTS: After 4 weeks on western diet, systemic adenoviral gene transfer was performed with hVEGF-A or control vectors. Effects on atherosclerotic lesion area and composition, lipoprotein profiles, and plasma lipoprotein lipase (LPL) activity were examined. On day 4, VEGF-A induced alterations in lipoprotein profiles and a significant negative correlation was observed between plasma LPL activity and VEGF-A levels. One month after gene transfer, no changes in atherosclerosis were observed in LDLR(-/-) and LDLR(-/-)ApoB(100/100) models, whereas both ApoE(-/-) models displayed increased en face lesion areas in thoracic and abdominal aortas. VEGF-A also reduced LPL mRNA in heart and white adipose tissue, whereas Angptl4 was increased, potentially providing further mechanistic explanation for the findings. CONCLUSION: VEGF-A gene transfer induced pro-atherogenic changes in lipoprotein profiles in all models. As a novel finding, VEGF-A also reduced LPL activity, which might underlie the observed changes in lipid profiles. However, VEGF-A was observed to increase atherosclerosis only in the ApoE(-/-) background, clearly indicating some mouse model-specific effects.


Assuntos
Aterosclerose/etiologia , Terapia Genética , Hiperlipidemias/terapia , Lipase Lipoproteica/metabolismo , Lipoproteínas/sangue , Fator A de Crescimento do Endotélio Vascular/genética , Adenoviridae/genética , Animais , Apolipoproteínas B/deficiência , Apolipoproteínas B/fisiologia , Modelos Animais de Doenças , Feminino , Hiperlipidemias/sangue , Hiperlipidemias/patologia , Lipídeos/sangue , Lipase Lipoproteica/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Receptores de LDL/fisiologia
2.
Cardiovasc Res ; 98(1): 107-15, 2013 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-23341579

RESUMO

AIMS: The loss of nuclear factor E2-related factor 2 (Nrf2) has been shown to protect against atherogenesis in apoE-deficient mice. The mechanism by which Nrf2 deficiency affords atheroprotection in this model is currently unknown, but combined systemic and local vascular effects on lesion macrophages have been proposed. We investigated the effect of bone marrow-specific loss of Nrf2 on early atherogenesis in low-density lipoprotein (LDL) receptor-deficient (LDLR(-/-)) mice, and assessed the effect of Nrf2 on cellular accumulation of modified LDLs and the expression of inflammatory markers in macrophages. METHODS AND RESULTS: The effect of bone marrow-specific loss of Nrf2 on atherogenesis was studied using bone marrow transplantation of wild-type (WT) or Nrf2(-/-) bone marrow to LDLR(-/-) mice. Mice transplanted with Nrf2(-/-) bone marrow and fed a high-fat diet for 6 weeks exhibited significantly larger atherosclerotic lesions than WT bone marrow transplanted mice. Moreover, in thioglycollate-elicited Nrf2(-/-) macrophages, the uptake of acetylated and malondialdehyde-modified LDLs was increased in comparison with WT controls, with the concomitant increase in the expression of scavenger receptor A and toll-like receptor 4. In addition, the expression of pro-inflammatory monocyte chemoattractant protein-1 and interleukin-6 were increased in Nrf2(-/-) vs. WT macrophages. CONCLUSION: Nrf2 deficiency specific to bone marrow-derived cells aggravates atherosclerosis in LDLR(-/-) mice. Furthermore, the loss of Nrf2 in macrophages enhances foam cell formation and promotes the pro-inflammatory phenotype.


Assuntos
Aterosclerose/etiologia , Macrófagos/fisiologia , Fator 2 Relacionado a NF-E2/fisiologia , Animais , Quimiocina CCL2/genética , Colesterol/metabolismo , Feminino , Camundongos , Camundongos Endogâmicos C57BL , Receptores de LDL/fisiologia , Receptores Depuradores/análise
3.
Atherosclerosis ; 225(2): 335-40, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-23102786

RESUMO

OBJECTIVE: Endothelial lipase (EL) regulates HDL cholesterol levels and in inflammatory states, like atherosclerosis, EL expression is increased contributing to low HDL cholesterol. The regulation of EL expression is poorly understood and has mainly been attributed to inflammatory stimuli. As sterol regulatory element binding proteins (SREBPs) are regulators of genes involved in lipid metabolism, we hypothesized that EL is regulated by SREBPs and that EL expression is modified by the SREBP activator vascular endothelial growth factor A (VEGF-A). METHODS: and results: Quantitative PCR and Western blot results demonstrated that starvation increased EL expression in human umbilical vein endothelial cells (HUVECs) and human aortic endothelial cells (HAECs). Also, 25-hydroxycholesterol (25HC), an inhibitor of SREBP activation inhibited EL expression. With siRNA-mediated inhibition of SREBPs the effect of starvation was shown to be SREBP-2 dependent. VEGF-A decreased EL expression in both endothelial cell lines used, most likely via inhibition of SREBP-2 binding determined by chromatin immunoprecipitation (ChIP). Furthermore, in atherosclerosis prone LDLR(-/-)ApoB(100/100) mice, systemic adenoviral gene transfer with human VEGF-A decreased EL mRNA in peripheral tissues and increased plasma HDL cholesterol. CONCLUSIONS: These results identify SREBPs as novel regulators of EL expression. VEGF-A as an endogenous EL inhibitor could be therapeutically relevant in atherosclerosis by increasing systemic HDL cholesterol levels.


Assuntos
HDL-Colesterol/sangue , Células Endoteliais/enzimologia , Lipase/metabolismo , Proteína de Ligação a Elemento Regulador de Esterol 2/metabolismo , Fator A de Crescimento do Endotélio Vascular/metabolismo , Adenoviridae/genética , Animais , Apolipoproteína B-100/deficiência , Apolipoproteína B-100/genética , Aterosclerose/sangue , Aterosclerose/enzimologia , Aterosclerose/genética , Sítios de Ligação , Western Blotting , Células Cultivadas , Imunoprecipitação da Cromatina , Modelos Animais de Doenças , Células Endoteliais/efeitos dos fármacos , Regulação Enzimológica da Expressão Gênica , Técnicas de Transferência de Genes , Vetores Genéticos , Células Endoteliais da Veia Umbilical Humana/efeitos dos fármacos , Células Endoteliais da Veia Umbilical Humana/enzimologia , Humanos , Hidroxicolesteróis/farmacologia , Lipase/genética , Camundongos , Camundongos Knockout , Reação em Cadeia da Polimerase , Regiões Promotoras Genéticas , Interferência de RNA , RNA Mensageiro/metabolismo , Receptores de LDL/deficiência , Receptores de LDL/genética , Proteína de Ligação a Elemento Regulador de Esterol 1/metabolismo , Proteína de Ligação a Elemento Regulador de Esterol 2/antagonistas & inibidores , Proteína de Ligação a Elemento Regulador de Esterol 2/genética , Fatores de Tempo , Transfecção , Fator A de Crescimento do Endotélio Vascular/genética
4.
J Gene Med ; 14(3): 182-90, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-22287362

RESUMO

BACKGROUND: Vascular endothelial growth factors (VEGFs) are central mediators in vascular development and lymphangiogenesis. VEGF-D contributes to the growth and formation of blood and lymphatic vessels, although its biological role is still somewhat unclear. METHODS: Transgenic mice, which express the mature form of human VEGF-D under endothelium-specific Tie1 promoter, were produced by the lentiviral perivitelline-injection method. The mice were followed up to generation F(5) and the effect of the transgene was analyzed. RESULTS: Transgenic mice had a high expression of human (h)VEGF-D in the endothelium in several tissues, such as kidney, liver, lung and spleen. However, transgenic mice developed tumors in lungs, kidneys, liver, mammary glands and lymph nodes upon aging and their mortality was also increased as a result of other pathological conditions. Hind limb ischemia was surgically induced in these mice and they were analyzed 1, 2 and 3 weeks after the ischemia operation. No significant differences were found in hVEGF-D mRNA expression, the number of capillaries or tissue repair between ischemic transgenic mice and transgene negative littermates. CONCLUSIONS: It is concluded that targeted unregulated long-term expression of hVEGF-D in endothelium may not be useful and reduces the life span of transgenic mice.


Assuntos
Endotélio/metabolismo , Membro Posterior/irrigação sanguínea , Isquemia/metabolismo , Neoplasias Experimentais/patologia , Transgenes/genética , Fator D de Crescimento do Endotélio Vascular/genética , Fator D de Crescimento do Endotélio Vascular/metabolismo , Análise de Variância , Animais , Vetores Genéticos/genética , Membro Posterior/patologia , Humanos , Imuno-Histoquímica , Lentivirus , Camundongos , Camundongos Transgênicos , Neoplasias Experimentais/genética , Regiões Promotoras Genéticas/genética , Receptor de TIE-1/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Taxa de Sobrevida
5.
JACC Cardiovasc Imaging ; 4(12): 1294-301, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-22172786

RESUMO

OBJECTIVES: This study investigated the effects of age, duration of a high-fat diet, and type 2 diabetes on atherosclerotic plaque development and uptake of (18)F-fluorodeoxyglucose ((18)F-FDG) in 2 mouse models. BACKGROUND: The animal's age and start time and duration of a high-fat diet have effects on plaque composition in atherosclerotic mice. METHODS: The aortas of atherosclerotic low-density lipoprotein receptor deficient mice expressing only apolipoprotein B100 (LDLR(-/-)ApoB(100/100)) and atherosclerotic and diabetic mice overexpressing insulin-like growth factor II (IGF-II/LDLR(-/-)ApoB(100/100)) were investigated at 4, 6, and 12 months of age and older after varying durations of high-fat diet. C57BL/6N mice on normal chow served as controls. Plaque size (intima-to-media ratio), macrophage density (Mac-3 staining), and plaque uptake of (18)F-FDG were studied by means of in vivo positron emission tomography/computed tomography by ex vivo autoradiography and by histological and immunohistochemical methods. RESULTS: From the ages of 4 to 6 months and 12 months and older, the plaque size increased and the macrophage density decreased. Compared with the controls, the in vivo imaging showed increased aortic (18)F-FDG uptake at 4 and 6 months, but not at 12 months and older. Autoradiography showed focal (18)F-FDG uptake in plaques at all time points (average plaque-to-normal vessel wall ratio: 2.4 ± 0.4, p < 0.001) with the highest uptake in plaques with high macrophage density. There were no differences in the plaque size, macrophage density, or uptake of (18)F-FDG between LDLR(-/-)ApoB(100/100) and IGF-II/LDLR(-/-)ApoB(100/100) mice at any time point. CONCLUSIONS: The 6-month-old LDLR(-/-)ApoB(100/100) and IGF-II/LDLR(-/-)ApoB(100/100) mice demonstrated highly inflamed, large, and extensive atherosclerotic plaques after 4 months of a high-fat diet, presenting a suitable model for studying the imaging of atherosclerotic plaque inflammation with (18)F-FDG. The presence of type 2 diabetes did not confound evaluation of plaque inflammation with (18)F-FDG.


Assuntos
Envelhecimento , Aterosclerose/diagnóstico por imagem , Aterosclerose/etiologia , Diabetes Mellitus Tipo 2/complicações , Dieta Hiperlipídica/efeitos adversos , Fluordesoxiglucose F18 , Imagem Multimodal , Placa Aterosclerótica/diagnóstico por imagem , Placa Aterosclerótica/etiologia , Tomografia por Emissão de Pósitrons , Compostos Radiofarmacêuticos , Tomografia Computadorizada por Raios X , Fatores Etários , Animais , Aorta/diagnóstico por imagem , Aorta/patologia , Aortografia , Apolipoproteína B-100/genética , Apolipoproteína B-100/metabolismo , Aterosclerose/genética , Aterosclerose/metabolismo , Aterosclerose/patologia , Autorradiografia , Diabetes Mellitus Tipo 2/genética , Diabetes Mellitus Tipo 2/metabolismo , Modelos Animais de Doenças , Fluordesoxiglucose F18/farmacocinética , Imuno-Histoquímica , Macrófagos/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Placa Aterosclerótica/genética , Placa Aterosclerótica/metabolismo , Placa Aterosclerótica/patologia , Compostos Radiofarmacêuticos/farmacocinética , Receptor IGF Tipo 2/genética , Receptor IGF Tipo 2/metabolismo , Receptores de LDL/deficiência , Receptores de LDL/genética , Fatores de Risco
6.
Arterioscler Thromb Vasc Biol ; 31(7): 1617-24, 2011 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-21474827

RESUMO

OBJECTIVE: The mature form of human vascular endothelial growth factor-D (hVEGF-D(ΔNΔC)) is an efficient angiogenic factor, but its full mechanism of action has remained unclear. We studied the effects of hVEGF-D(ΔNΔC) in endothelial cells using gene array, signaling, cell culture, and in vivo gene transfer techniques. METHODS AND RESULTS: Concomitant with the angiogenic and proliferative responses, hVEGF-D(ΔNΔC) enhanced the phosphorylation of VEGF receptor-2, Akt, and endothelial nitric oxide synthase. Gene arrays, quantitative reverse transcription-polymerase chain reaction, and Western blot revealed increases in VEGF-A, stanniocalcin-1 (STC1), and neuropilin (NRP) 2 expression by hVEGF-D(ΔNΔC) stimulation, whereas induction with hVEGF-A(165) altered the expression of STC1 and NRP1, another coreceptor for VEGFs. The effects of hVEGF-D(ΔNΔC) were seen only under high-serum conditions, whereas for hVEGF-A(165), the strongest response was observed under low-serum conditions. The hVEGF-D(ΔNΔC)-induced upregulation of STC1 and NRP2 was also evident in vivo in mouse skeletal muscle treated with hVEGF-D(ΔNΔC) by adenoviral gene delivery. The importance of NRP2 in hVEGF-D(ΔNΔC) signaling was further studied with NRP2 small interfering RNA and NRP antagonist, which were able to block hVEGF-D(ΔNΔC)-induced survival of endothelial cells. CONCLUSIONS: In this study, the importance of serum and upregulation of NRP2 and STC1 for VEGF-D(ΔNΔC) effects were demonstrated. Better knowledge of VEGF-D(ΔNΔC) signaling and regulation is valuable for the development of efficient and safe VEGF-D(ΔNΔC)-based therapeutic applications for cardiovascular diseases.


Assuntos
Células Endoteliais/metabolismo , Glicoproteínas/metabolismo , Músculo Esquelético/irrigação sanguínea , Neovascularização Fisiológica , Neuropilina-2/metabolismo , Fator A de Crescimento do Endotélio Vascular/metabolismo , Fator D de Crescimento do Endotélio Vascular/metabolismo , Animais , Apolipoproteína B-100/deficiência , Apolipoproteína B-100/genética , Western Blotting , Proliferação de Células , Células Cultivadas , Perfilação da Expressão Gênica , Membro Posterior , Humanos , Camundongos , Camundongos Knockout , Neuropilina-1/metabolismo , Neuropilina-2/genética , Óxido Nítrico Sintase Tipo III/metabolismo , Fosforilação , Proteínas Proto-Oncogênicas c-akt/metabolismo , Interferência de RNA , Receptores de LDL/deficiência , Receptores de LDL/genética , Proteínas Recombinantes/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais , Fatores de Tempo , Transdução Genética , Regulação para Cima , Fator D de Crescimento do Endotélio Vascular/genética , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo
7.
Cardiovasc Res ; 88(3): 530-8, 2010 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-20634212

RESUMO

AIMS: Macrophage scavenger receptor A (SR-A) and class B scavenger receptor CD36 (CD36) contribute to foam cell formation and atherogenesis via uptake of modified lipoproteins. So far, the role of these scavenger receptors has been studied mainly using knockout models totally lacking these receptors. We studied the role of SR-A and CD36 in foam cell formation and atherogenesis by RNA interference (RNAi)-mediated silencing, which is a clinically feasible method to down-regulate the expression of these receptors. METHODS AND RESULTS: We constructed lentivirus vectors encoding short hairpin RNAs (shRNAs) against mouse SR-A and CD36. Decreased SR-A but not CD36 expression led to reduced foam cell formation caused by acetylated low-density lipoprotein (LDL) in mouse macrophages, whereas the uptake of oxidized LDL was not altered. More importantly, silencing of SR-A upregulates CD36 and vice versa. In LDL receptor-deficient apolipoprotein B100 (LDLR(-/-)ApoB(100/100)) mice kept on a western diet, silencing of either SR-A or CD36 in bone marrow cells led to a marked decrease (37.4 and 34.2%, respectively) in cross-sectional lesion area, whereas simultaneous silencing of both receptors was not effective. CONCLUSION: Our results suggest that silencing of either SR-A or CD36 alone reduces atherogenesis in mice. However, due to reciprocal upregulation, silencing of both SR-A and CD36 is not effective.


Assuntos
Aterosclerose/prevenção & controle , Antígenos CD36/genética , Hiperlipidemias/complicações , Interferência de RNA , Receptores Depuradores Classe A/genética , Regulação para Cima/genética , Animais , Apolipoproteínas B/genética , Apolipoproteínas B/metabolismo , Aterosclerose/genética , Aterosclerose/metabolismo , Antígenos CD36/metabolismo , Linhagem Celular , Modelos Animais de Doenças , Feminino , Células Espumosas/metabolismo , Células Espumosas/patologia , Humanos , Hiperlipidemias/metabolismo , Hiperlipidemias/patologia , Lentivirus/genética , Macrófagos/metabolismo , Macrófagos/patologia , Masculino , Camundongos , Camundongos Knockout , Receptores de LDL/genética , Receptores de LDL/metabolismo , Receptores Depuradores Classe A/metabolismo
8.
Cardiovasc Res ; 86(1): 122-30, 2010 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-19955220

RESUMO

AIMS: Vascular endothelial growth factors (VEGFs) and their receptors (VEGF-Rs) are among the most powerful factors regulating vascular growth. However, it has remained unknown whether stimulation of VEGF-R1, VEGF-R2 or both of the receptors produces the best angiogenic responses in myocardium. The aim of this study was to compare the VEGF-R1-specific ligand VEGF-B(186), VEGF-R2-specific ligand VEGF-E and VEGF-A(165,) which stimulates both receptors, regarding their effects on angiogenesis and left ventricular function in mice. METHODS AND RESULTS: High-resolution echocardiography was used to guide the closed-chest injections of adenoviral (Ad) vectors expressing VEGF-B(186,) VEGF-E, and VEGF-A(165) into the anterior wall of the left ventricle in C57Bl/6J mice. Angiogenic and functional effects were analysed using histology, ultrasound and perfusion analyses 6 (D6) and 14 (D14) days after the Ad injection. AdVEGF-A(165) induced a strong angiogenic response seen as an enlargement of myocardial capillaries whereas angiogenesis induced by AdVEGF-B(186) and AdVEGF-E seemed more physiological. The increase in the capillary area was accompanied with an increase in myocardial perfusion at D6 after the gene injection. AdVEGF-A(165) and AdVEGF-E induced endothelial-specific proliferation whereas AdVEGF-B(186) mostly induced proliferation of cardiomyocytes. AdVEGF-A(165) induced more pronounced tissue damage than AdVEGF-B(186) and AdVEGF-E. Left ventricular function measured as ejection fraction did not change during the follow-up. AdVEGF-A(165) increased both VEGF-R1 and VEGF-R2 protein expression whereas AdVEGF-B(186) and AdVEGF-E did not affect endogenous receptor expression levels. CONCLUSION: AdVEGF-B(186) and AdVEGF-E are equally potent in inducing therapeutic angiogenesis in mouse myocardium and produce less side effects than AdVEGF-A(165).


Assuntos
Neovascularização Fisiológica/fisiologia , Fator A de Crescimento do Endotélio Vascular/metabolismo , Receptor 1 de Fatores de Crescimento do Endotélio Vascular/metabolismo , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo , Função Ventricular Esquerda/fisiologia , Adenoviridae/genética , Animais , Western Blotting , Divisão Celular/efeitos dos fármacos , Divisão Celular/fisiologia , Circulação Coronária/efeitos dos fármacos , Circulação Coronária/fisiologia , Ecocardiografia , Ensaio de Imunoadsorção Enzimática , Ligantes , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Modelos Cardiovasculares , Miocardite/induzido quimicamente , Miocardite/metabolismo , Miocardite/fisiopatologia , Neovascularização Fisiológica/efeitos dos fármacos , Fragmentos de Peptídeos/genética , Fragmentos de Peptídeos/metabolismo , Fragmentos de Peptídeos/farmacologia , Volume Sistólico/efeitos dos fármacos , Volume Sistólico/fisiologia , Fator A de Crescimento do Endotélio Vascular/genética , Fator A de Crescimento do Endotélio Vascular/farmacologia , Fator B de Crescimento do Endotélio Vascular/genética , Fator B de Crescimento do Endotélio Vascular/metabolismo , Fator B de Crescimento do Endotélio Vascular/farmacologia , Função Ventricular Esquerda/efeitos dos fármacos , Proteínas Virais/genética , Proteínas Virais/metabolismo , Proteínas Virais/farmacologia
9.
Circ Res ; 105(6): 604-9, 2009 Sep 11.
Artigo em Inglês | MEDLINE | ID: mdl-19696410

RESUMO

RATIONALE: We studied a possibility that shRNAs can lead to transcriptional gene activation at the promoter level via epigenetic mechanism. OBJECTIVE: The purpose of this study was to test the effects on vascular endothelial growth factor (VEGF-A) expression by promoter targeted small hairpin RNAs (shRNAs) in vitro and in experimental animals in vivo using stable local lentiviral gene transfer. METHODS AND RESULTS: One shRNA was identified which strongly increased VEGF-A expression in C166 endothelial cells at mRNA and protein level whereas another shRNA decreased VEGF-A expression. Quantitative chromatin immunoprecipitation analysis revealed that the repressing shRNA caused epigenetic changes, which increased nucleosome density within the promoter and transcription start site and led to repression of VEGF-A expression. Epigenetic changes caused by the activating shRNA were opposite to those caused by the repressing shRNA. These results were confirmed in vivo in an ischemic mouse hindlimb model after local gene transfer where VEGF-A upregulation achieved by promoter-targeted shRNA increased vascularity and blood flow. CONCLUSIONS: We show that lentivirus-mediated delivery of shRNA molecules targeted to specific regions in the mVEGF-A promoter either induce or repress VEGF-A expression via epigenetic modulation. Thus, we describe a new approach of gene therapy, epigenetherapy, based on an epigenetic mechanism at the promoter level. Controlling transcription through manipulation of specific epigenetic marks provides a novel approach for the treatment of several diseases.


Assuntos
Epigênese Genética , Terapia Genética/métodos , Membro Posterior/irrigação sanguínea , Isquemia/terapia , Lentivirus , Regiões Promotoras Genéticas , RNA/biossíntese , Fator A de Crescimento do Endotélio Vascular/genética , Animais , Linhagem Celular , Células Endoteliais/metabolismo , Isquemia/genética , Camundongos , RNA/genética , Transcrição Gênica , Fator A de Crescimento do Endotélio Vascular/metabolismo
10.
Circ Res ; 103(1): e1-9, 2008 Jul 03.
Artigo em Inglês | MEDLINE | ID: mdl-18535259

RESUMO

Besides their well-characterized proinflammatory and proatherogenic effects, oxidized phospholipids, such as oxPAPC (oxidized 1-palmitoyl-2-arachidonoyl-sn-glycero-phosphocholine) have been shown to have beneficial responses in vascular cells via induction of antioxidant enzymes such as heme oxygenase-1. We therefore hypothesized that oxPAPC could evoke a general cytoprotective response via activation of antioxidative transcription factor Nrf2. Here, we show that oxPAPC increases nuclear accumulation of Nrf2. Using the small interfering RNA approach, we demonstrate that Nrf2 is critical in mediating the induction of glutamate-cysteine ligase modifier subunit (GCLM) and NAD(P)H quinone oxidoreductase-1 (NQO1) by oxPAPC in human endothelial cells, whereas the contribution to the induction of heme oxygenase-1 was less significant. The induction of GCLM and NQO1 was attenuated by reduction of electrophilic groups with sodium borohydrate, as well as treatment with thiol antioxidant N-acetylcysteine, suggesting that the thiol reactivity of oxPAPC is largely mediating its effect on Nrf2-responsive genes. Moreover, we show that oxidized phospholipid having a highly electrophilic isoprostane ring in its sn-2 position is a potent inducer of Nrf2 target genes. Finally, we demonstrate that the oxPAPC-inducible expression of heme oxygenase-1, GCLM, and NQO1 is lower in Nrf2-null than wild-type mouse carotid arteries in vivo. We suggest that the activation of Nrf2 by oxidized phospholipids provides a mechanism by which their deleterious effects are limited in the vasculature.


Assuntos
Antioxidantes/metabolismo , Artérias Carótidas/enzimologia , Núcleo Celular/metabolismo , Células Endoteliais/enzimologia , Regulação Enzimológica da Expressão Gênica/efeitos dos fármacos , Fator 2 Relacionado a NF-E2/metabolismo , Fosfatidilcolinas/farmacocinética , Transporte Ativo do Núcleo Celular/efeitos dos fármacos , Transporte Ativo do Núcleo Celular/fisiologia , Animais , Artérias Carótidas/citologia , Núcleo Celular/genética , Células Endoteliais/citologia , Regulação Enzimológica da Expressão Gênica/fisiologia , Glutamato-Cisteína Ligase/biossíntese , Glutamato-Cisteína Ligase/genética , Heme Oxigenase-1/biossíntese , Heme Oxigenase-1/genética , Camundongos , Camundongos Mutantes , NAD(P)H Desidrogenase (Quinona) , NADPH Desidrogenase/biossíntese , NADPH Desidrogenase/genética , Fator 2 Relacionado a NF-E2/antagonistas & inibidores , Fator 2 Relacionado a NF-E2/genética , Oxirredução/efeitos dos fármacos , Fosfatidilcolinas/metabolismo , RNA Interferente Pequeno/genética
11.
Circ Res ; 101(10): 1058-67, 2007 Nov 09.
Artigo em Inglês | MEDLINE | ID: mdl-17872464

RESUMO

No mouse model is currently available where the induction of type 2 diabetes on an atherosclerotic background could be achieved without significant concomitant changes in plasma lipid levels. We crossbred 2 genetically modified mouse strains to achieve a model expressing both atherosclerosis and characteristics of type 2 diabetes. For atherosclerotic background we used low-density lipoprotein receptor-deficient mice synthetizing only apolipoprotein B100 (LDLR(-/-) ApoB(100/100)). Diabetic background was obtained from transgenic mice overexpressing insulin-like growth factor-II (IGF-II) in pancreatic beta cells. Thorough phenotypic characterization was performed in 6- and 15-month-old mice on both normal and high-fat Western diet. Results indicated that IGF-II transgenic LDLR(-/-)ApoB(100/100) mice demonstrated insulin resistance, hyperglycemia, and mild hyperinsulinemia compared with hypercholesterolemic LDLR(-/-)ApoB(100/100) controls. In addition, old IGF-II/LDLR(-/-)ApoB(100/100) mice displayed significantly increased lesion calcification, which was more related to insulin resistance than glucose levels, and significantly higher baseline expression in aorta of several genes related to calcification and inflammation. Lipid levels of IGF-II/LDLR(-/-)ApoB(100/100) mice did not differ from LDLR(-/-)ApoB(100/100) controls at any time. In conclusion, type 2 diabetic factors induce increased calcification and lesion progression without any lipid changes in a new mouse model of diabetic macroangiopathy.


Assuntos
Calcinose/complicações , Angiopatias Diabéticas/complicações , Modelos Animais de Doenças , Hipercolesterolemia/complicações , Hiperglicemia/complicações , Camundongos Transgênicos , Animais , Apolipoproteínas B/genética , Aterosclerose/complicações , Aterosclerose/patologia , Aterosclerose/fisiopatologia , Glicemia , Calcinose/patologia , Calcinose/fisiopatologia , Diabetes Mellitus Tipo 2/complicações , Diabetes Mellitus Tipo 2/patologia , Diabetes Mellitus Tipo 2/fisiopatologia , Angiopatias Diabéticas/patologia , Angiopatias Diabéticas/fisiopatologia , Hipercolesterolemia/patologia , Hipercolesterolemia/fisiopatologia , Hiperglicemia/patologia , Hiperglicemia/fisiopatologia , Resistência à Insulina , Fator de Crescimento Insulin-Like II/genética , Lipídeos/sangue , Camundongos , Camundongos Endogâmicos C57BL , Osteoprotegerina/sangue , Receptores de LDL/genética
12.
Acta Obstet Gynecol Scand ; 86(9): 1035-40, 2007.
Artigo em Inglês | MEDLINE | ID: mdl-17712641

RESUMO

BACKGROUND: The options for destroying the full thickness of endometrium are very limited, relying mainly on surgical endometrial ablation. A nonsurgical, safe method would benefit many women in clinical settings. This study was undertaken to investigate the potential of gene therapy to transfect and destroy endometriun at different stages of the reproductive cycle in rabbits. METHODS: We used adenoviruses carrying the LacZ (AdvLacZ) marker gene, indicating the transfection efficiency, and adenoviruses carrying thymidine kinase (AdvTK) followed by intravenous ganciclovir therapy as a potential treatment for excess endometrial growth. Ganciclovir was given intravenously after AdvTK treatment. Adenoviruses were injected intraluminally into the uterus of nonpregnant, pseudopregnant, and pregnant New Zealand White rabbits (n=25). RESULTS: After AdvLacZ gene transfer into the uterus of intact rabbits, transduced cells were observed in uterine muscle and endometrial stroma. In pseudopregnant and pregnant rabbits the endometrium was proliferative and transduction was restricted to endometrial epithelium. However, no treatment effect was observed after AdvTK gene therapy although the transduction with AdvLacZ was clearly detectable. CONCLUSIONS: It is concluded that the transduction pattern of uterine tissues varies significantly with the reproductive cycle. Secondly, although the transduction efficiency was relatively high in the endometrial epithelium, the effect of the AdvTK and ganciclovir treatment was poor and not sufficient for clinical applications.


Assuntos
Adenoviridae/genética , Endométrio/patologia , Terapia Genética/métodos , Óperon Lac/genética , Timidina Quinase/genética , Transdução Genética/métodos , Adenoviridae/enzimologia , Animais , Antivirais/farmacologia , Apoptose , Endométrio/enzimologia , Ciclo Estral/fisiologia , Feminino , Ganciclovir/farmacologia , Expressão Gênica , Vetores Genéticos , Reação em Cadeia da Polimerase , Gravidez , Coelhos , Transfecção/métodos , Útero/metabolismo , Útero/patologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA