Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Angiogenesis ; 25(1): 129-143, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-34432198

RESUMO

Vein grafting is a frequently used surgical intervention for cardiac revascularization. However, vein grafts display regions with intraplaque (IP) angiogenesis, which promotes atherogenesis and formation of unstable plaques. Graft neovessels are mainly composed of endothelial cells (ECs) that largely depend on glycolysis for migration and proliferation. In the present study, we aimed to investigate whether loss of the glycolytic flux enzyme phosphofructokinase-2/fructose-2,6-bisphosphatase 3 (PFKFB3) in ECs inhibits IP angiogenesis and as such prevents unstable plaque formation. To this end, apolipoprotein E deficient (ApoE-/-) mice were backcrossed to a previously generated PFKFB3fl/fl Cdh5iCre mouse strain. Animals were injected with either corn oil (ApoE-/-PFKFB3fl/fl) or tamoxifen (ApoE-/-PFKFB3ECKO), and were fed a western-type diet for 4 weeks prior to vein grafting. Hereafter, mice received a western diet for an additional 28 days and were then sacrificed for graft assessment. Size and thickness of vein graft lesions decreased by 35 and 32%, respectively, in ApoE-/-PFKFB3ECKO mice compared to controls, while stenosis diminished by 23%. Moreover, vein graft lesions in ApoE-/-PFKFB3ECKO mice showed a significant reduction in macrophage infiltration (29%), number of neovessels (62%), and hemorrhages (86%). EC-specific PFKFB3 deletion did not show obvious adverse effects or changes in general metabolism. Interestingly, RT-PCR showed an increased M2 macrophage signature in vein grafts from ApoE-/-PFKFB3ECKO mice. Altogether, EC-specific PFKFB3 gene deletion leads to a significant reduction in lesion size, IP angiogenesis, and hemorrhagic complications in vein grafts. This study demonstrates that inhibition of endothelial glycolysis is a promising therapeutic strategy to slow down plaque progression.


Assuntos
Células Endoteliais , Neovascularização Patológica , Fosfofrutoquinase-2/genética , Animais , Modelos Animais de Doenças , Células Endoteliais/metabolismo , Deleção de Genes , Glicólise , Camundongos , Neovascularização Patológica/genética , Fosfofrutoquinase-2/metabolismo
2.
Arterioscler Thromb Vasc Biol ; 40(5): 1168-1181, 2020 05.
Artigo em Inglês | MEDLINE | ID: mdl-32188275

RESUMO

OBJECTIVE: Intraplaque neovascularization is an important feature of unstable human atherosclerotic plaques. However, its impact on plaque formation and stability is poorly studied. Because proliferating endothelial cells generate up to 85% of their ATP from glycolysis, we investigated whether pharmacological inhibition of glycolytic flux by the small-molecule 3PO (3-[3-pyridinyl]-1-[4-pyridinyl]-2-propen-1-one) could have beneficial effects on plaque formation and composition. Approach and Results: ApoE-/- (apolipoprotein E deficient) mice treated with 3PO (50 µg/g, ip; 4×/wk, 4 weeks) showed a metabolic switch toward ketone body formation. Treatment of ApoE-/-Fbn1C1039G+/- mice with 3PO (50 µg/g, ip) either after 4 (preventive, twice/wk, 10 weeks) or 16 weeks of Western diet (curative, 4×/wk, 4 weeks) inhibited intraplaque neovascularization by 50% and 38%, respectively. Plaque formation was significantly reduced in all 3PO-treated animals. This effect was independent of intraplaque neovascularization. In vitro experiments showed that 3PO favors an anti-inflammatory M2 macrophage subtype and suppresses an M1 proinflammatory phenotype. Moreover, 3PO induced autophagy, which in turn impaired NF-κB (nuclear factor-kappa B) signaling and inhibited TNF-α (tumor necrosis factor-alpha)-mediated VCAM-1 (vascular cell adhesion molecule-1) and ICAM-1 (intercellular adhesion molecule-1) upregulation. Consistently, a preventive 3PO regimen reduced endothelial VCAM-1 expression in vivo. Furthermore, 3PO improved cardiac function in ApoE-/-Fbn1C1039G+/- mice after 10 weeks of treatment. CONCLUSIONS: Partial inhibition of glycolysis restrained intraplaque angiogenesis without affecting plaque composition. However, less plaques were formed, which was accompanied by downregulation of endothelial adhesion molecules-an event that depends on autophagy induction. Inhibition of coronary plaque formation by 3PO resulted in an overall improved cardiac function.


Assuntos
Artérias/efeitos dos fármacos , Aterosclerose/tratamento farmacológico , Glicólise/efeitos dos fármacos , Neovascularização Patológica , Placa Aterosclerótica , Piridinas/farmacologia , Animais , Artérias/metabolismo , Artérias/patologia , Aterosclerose/genética , Aterosclerose/metabolismo , Aterosclerose/patologia , Autofagia/efeitos dos fármacos , Células Cultivadas , Modelos Animais de Doenças , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/metabolismo , Células Endoteliais/patologia , Feminino , Fibrilina-1/genética , Fibrilina-1/metabolismo , Humanos , Molécula 1 de Adesão Intercelular/metabolismo , Macrófagos/efeitos dos fármacos , Macrófagos/metabolismo , Macrófagos/patologia , Camundongos Knockout para ApoE , NF-kappa B/metabolismo , Fenótipo , Molécula 1 de Adesão de Célula Vascular/metabolismo
3.
J Vasc Res ; 57(6): 348-354, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32610324

RESUMO

Multiple lines of evidence suggest that intraplaque (IP) neovascularization promotes atherosclerotic plaque growth, destabilization, and rupture. However, pharmacological inhibition of IP neovascularization remains largely unexplored due to the limited number of animal models that develop IP neovessels and the lack of reliable methods for visualizing IP angiogenesis. Here, we applied 3D confocal microscopy with an optimized tissue-clearing process, immunolabeling-enabled three-dimensional imaging of solvent-cleared organs, to visualize IP neovessels in apolipoprotein E-deficient (ApoE-/-) mice carrying a heterozygous mutation (C1039+/-) in the fibrillin-1 gene. Unlike regular ApoE-/- mice, this mouse model is characterized by the presence of advanced plaques with evident IP neovascularization. Plaques were stained with antibodies against endothelial marker CD31 for 3 days, followed by incubation with fluorescently labeled secondary antibodies. Subsequent tissue clearing with dichloromethane (DCM)/methanol, DCM, and dibenzyl ether allowed easy visualization and 3D reconstruction of the IP vascular network while plaque morphology remained intact.


Assuntos
Artérias Carótidas/patologia , Doenças das Artérias Carótidas/patologia , Imageamento Tridimensional , Microscopia Confocal , Neovascularização Patológica , Placa Aterosclerótica , Animais , Biomarcadores/metabolismo , Complexo CD3/metabolismo , Artérias Carótidas/metabolismo , Doenças das Artérias Carótidas/genética , Doenças das Artérias Carótidas/metabolismo , Modelos Animais de Doenças , Feminino , Fibrilina-1/genética , Fibrilina-1/metabolismo , Técnica Indireta de Fluorescência para Anticorpo , Camundongos Knockout para ApoE , Mutação
4.
Proc Natl Acad Sci U S A ; 112(41): 12812-7, 2015 Oct 13.
Artigo em Inglês | MEDLINE | ID: mdl-26417068

RESUMO

The contribution of endothelial-derived miR-17∼92 to ischemia-induced arteriogenesis has not been investigated in an in vivo model. In the present study, we demonstrate a critical role for the endothelial-derived miR-17∼92 cluster in shaping physiological and ischemia-triggered arteriogenesis. Endothelial-specific deletion of miR-17∼92 results in an increase in collateral density limbs and hearts and in ischemic limbs compared with control mice, and consequently improves blood flow recovery. Individual cluster components positively or negatively regulate endothelial cell (EC) functions in vitro, and, remarkably, ECs lacking the cluster spontaneously form cords in a manner rescued by miR-17a, -18a, and -19a. Using both in vitro and in vivo analyses, we identified FZD4 and LRP6 as targets of miR-19a/b. Both of these targets were up-regulated in 17∼92 KO ECs compared with control ECs, and both were shown to be targeted by miR-19 using luciferase assays. We demonstrate that miR-19a negatively regulates FZD4, its coreceptor LRP6, and WNT signaling, and that antagonism of miR-19a/b in aged mice improves blood flow recovery after ischemia and reduces repression of these targets. Collectively, these data provide insights into miRNA regulation of arterialization and highlight the importance of vascular WNT signaling in maintaining arterial blood flow.


Assuntos
Receptores Frizzled/metabolismo , Proteína-6 Relacionada a Receptor de Lipoproteína de Baixa Densidade/metabolismo , MicroRNAs/metabolismo , Família Multigênica/fisiologia , Neovascularização Fisiológica/fisiologia , Via de Sinalização Wnt/fisiologia , Animais , Receptores Frizzled/genética , Isquemia/genética , Isquemia/metabolismo , Isquemia/patologia , Proteína-6 Relacionada a Receptor de Lipoproteína de Baixa Densidade/genética , Camundongos , Camundongos Knockout , MicroRNAs/genética
5.
ACS Med Chem Lett ; 11(5): 933-939, 2020 May 14.
Artigo em Inglês | MEDLINE | ID: mdl-32435408

RESUMO

PFKFB3, a glycolysis-related enzyme upregulated in inflammatory conditions and angiogenesis, is an emerging target for diagnosis and therapy of atherosclerosis. The fluorinated phenoxindazole [18F]ZCDD083 was synthesized, radiolabeled in 17 ± 5% radiochemical yield and >99% radiochemical purity, and formulated for preclinical PET/CT imaging in mice. In vivo stability analysis showed no significant metabolite formation. Biodistribution studies showed high blood pool activity and slow hepatobiliary clearance. Significant activity was detected in the lung 2 h postinjection (pi) (11.0 ± 1.5%ID/g), while at 6 h pi no pulmonary background was observed. Ex vivo autoradiography at 6 h pi showed significant high uptake of [18F]ZCDD083 in the arch region and brachiocephalic artery of atherosclerotic mice, and no uptake in control mice, matching plaques distribution seen by lipid staining along with PFKFB3 expression seen by immunofluorescent staining. In vivo PET scans showed higher aortic region uptake of [18F]ZCDD083 in atherosclerotic ApoE-/-Fbn1C1039G+/- than in control mice (0.78 ± 0.05 vs 0.44 ± 0.09%ID/g). [18F]ZCDD083 was detected in aortic arch and brachiocephalic artery of ApoE-/- (with moderate atherosclerosis) and ApoE-/-Fbn1C1039G+/- (with severe, advanced atherosclerosis) mice, suggesting this tracer may be useful for the noninvasive detection of atherosclerotic plaques in vivo.

6.
FEBS Lett ; 594(18): 3067-3075, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32620030

RESUMO

6-Phosphofructo-2-kinase/fructose-2,6-bisphosphatase isoform 3 (PFKFB3) is a key enzyme of the glycolytic pathway, and it plays an essential role in angiogenesis. 3-(3-Pyridinyl)-1-(4-pyridinyl)-2-propen-1-one (3PO) is frequently used as a glycolysis inhibitor and is thought to inhibit PFKFB3. However, this latter effect of 3PO has never been investigated in detail and was the aim of the present study. To demonstrate binding of 3PO to PFKFB3, we used isothermal titration calorimetry. However, 3PO did not bind to PFKFB3, even up to 750 µm, in contrast to 3 µm of AZ67, which is a potent and specific PFKFB3 inhibitor. Instead, 3PO accumulated lactic acid inside the cells, leading to a decrease in the intracellular pH and an inhibition of enzymatic reactions of the glycolytic pathway.


Assuntos
Glicólise/efeitos dos fármacos , Células Endoteliais da Veia Umbilical Humana/metabolismo , Fosfofrutoquinase-2/metabolismo , Piridinas/farmacologia , Humanos , Ligação Proteica
7.
JCI Insight ; 5(3)2020 02 13.
Artigo em Inglês | MEDLINE | ID: mdl-32051336

RESUMO

Vascular inflammation is present in many cardiovascular diseases, and exogenous glucocorticoids have traditionally been used as a therapy to suppress inflammation. However, recent data have shown that endogenous glucocorticoids, acting through the endothelial glucocorticoid receptor, act as negative regulators of inflammation. Here, we performed ChIP for the glucocorticoid receptor, followed by next-generation sequencing in mouse endothelial cells to investigate how the endothelial glucocorticoid receptor regulates vascular inflammation. We identified a role of the Wnt signaling pathway in this setting and show that loss of the endothelial glucocorticoid receptor results in upregulation of Wnt signaling both in vitro and in vivo using our validated mouse model. Furthermore, we demonstrate glucocorticoid receptor regulation of a key gene in the Wnt pathway, Frzb, via a glucocorticoid response element gleaned from our genomic data. These results suggest a role for endothelial Wnt signaling modulation in states of vascular inflammation.


Assuntos
Endotélio Vascular/metabolismo , Receptores de Glucocorticoides/metabolismo , Via de Sinalização Wnt , Animais , Imunoprecipitação da Cromatina , DNA/metabolismo , Dexametasona/farmacologia , Endotélio Vascular/citologia , Peptídeos e Proteínas de Sinalização Intracelular/genética , Masculino , Camundongos , Camundongos Knockout , Família Multigênica , Ligação Proteica , Receptores de Glucocorticoides/efeitos dos fármacos , Vasculite/metabolismo
8.
Vascul Pharmacol ; 112: 72-78, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-29933080

RESUMO

Atherosclerosis is a complex multifactorial disease that affects large and medium-sized arteries. Rupture of atherosclerotic plaques and subsequent acute cardiovascular complications remain a leading cause of death and morbidity in the Western world. There is a considerable difference in safety profile between a stable and a vulnerable, rupture-prone lesion. The need for plaque-stabilizing therapies is high, and for a long time the lack of a suitable animal model mimicking advanced human atherosclerotic plaques made it very difficult to make progress in this area. Evidence from human plaques indicates that intra-plaque (IP) angiogenesis promotes atherosclerosis and plaque destabilization. Although neovascularization has been widely investigated in cancer, studies on the pharmacological inhibition of this phenomenon in atherosclerosis are scarce, mainly due to the lack of an appropriate animal model. By using ApoE-/- Fbn1C1039G+/- mice, a novel model of vulnerable plaques, we were able to investigate the effect of pharmacological inhibition of various mechanisms of IP angiogenesis on plaque destabilization and atherogenesis. In the present review, we discuss the following potential pharmacological strategies to inhibit IP angiogenesis: (1) inhibition of vascular endothelial growth factor signalling, (2) inhibition of glycolytic flux, and (3) inhibition of fatty acid oxidation. On the long run, IP neovascularization might be applicable as a therapeutic target to induce plaque stabilization on top of lipid-lowering treatment.


Assuntos
Inibidores da Angiogênese/farmacologia , Artérias/efeitos dos fármacos , Aterosclerose/tratamento farmacológico , Neovascularização Patológica , Placa Aterosclerótica , Animais , Artérias/metabolismo , Artérias/patologia , Aterosclerose/metabolismo , Aterosclerose/patologia , Modelos Animais de Doenças , Quimioterapia Combinada , Ácidos Graxos/metabolismo , Glicólise/efeitos dos fármacos , Humanos , Hipolipemiantes/farmacologia , Oxirredução , Receptores de Fatores de Crescimento do Endotélio Vascular/antagonistas & inibidores , Receptores de Fatores de Crescimento do Endotélio Vascular/metabolismo , Transdução de Sinais/efeitos dos fármacos , Fator A de Crescimento do Endotélio Vascular/antagonistas & inibidores , Fator A de Crescimento do Endotélio Vascular/metabolismo
9.
Eur J Pharmacol ; 816: 3-13, 2017 Dec 05.
Artigo em Inglês | MEDLINE | ID: mdl-28483459

RESUMO

An ideal animal model of atherosclerosis resembles human anatomy and pathophysiology and has the potential to be used in medical and pharmaceutical research to obtain results that can be extrapolated to human medicine. Moreover, it must be easy to acquire, can be maintained at a reasonable cost, is easy to handle and shares the topography of the lesions with humans. In general, animal models of atherosclerosis are based on accelerated plaque formation due to a cholesterol-rich/Western-type diet, manipulation of genes involved in the cholesterol metabolism, and the introduction of additional risk factors for atherosclerosis. Mouse and rabbit models have been mostly used, followed by pigs and non-human primates. Each of these models has its advantages and limitations. The mouse has become the predominant species to study experimental atherosclerosis because of its rapid reproduction, ease of genetic manipulation and its ability to monitor atherogenesis in a reasonable time frame. Both Apolipoprotein E deficient (ApoE-/-) and LDL-receptor (LDLr) knockout mice have been frequently used, but also ApoE/LDLr double-knockout, ApoE3-Leiden and PCSK9-AAV mice are valuable tools in atherosclerosis research. However, a great challenge was the development of a model in which intra-plaque microvessels, haemorrhages, spontaneous atherosclerotic plaque ruptures, myocardial infarction and sudden death occur consistently. These features are present in ApoE-/-Fbn1C1039G+/- mice, which can be used as a validated model in pre-clinical studies to evaluate novel plaque-stabilizing drugs.


Assuntos
Aterosclerose , Modelos Animais de Doenças , Animais , Aterosclerose/genética , Aterosclerose/metabolismo , Aterosclerose/patologia , Placa Aterosclerótica/genética , Placa Aterosclerótica/metabolismo , Placa Aterosclerótica/patologia
10.
EMBO Mol Med ; 8(6): 643-53, 2016 06.
Artigo em Inglês | MEDLINE | ID: mdl-27137489

RESUMO

Abnormal remodeling of atherosclerotic plaques can lead to rupture, acute myocardial infarction, and death. Enhancement of plaque extracellular matrix (ECM) may improve plaque morphology and stabilize lesions. Here, we demonstrate that chronic administration of LNA-miR-29 into an atherosclerotic mouse model improves indices of plaque morphology. This occurs due to upregulation of miR-29 target genes of the ECM (col1A and col3A) resulting in reduced lesion size, enhanced fibrous cap thickness, and reduced necrotic zones. Sustained LNA-miR-29 treatment did not affect circulating lipids, blood chemistry, or ECM of solid organs including liver, lung, kidney, spleen, or heart. Collectively, these data support the idea that antagonizing miR-29 may promote beneficial plaque remodeling as an independent approach to stabilize vulnerable atherosclerotic lesions.


Assuntos
Aterosclerose/patologia , MicroRNAs/antagonistas & inibidores , Animais , Colágeno Tipo I/metabolismo , Cadeia alfa 1 do Colágeno Tipo I , Colágeno Tipo III/metabolismo , Modelos Animais de Doenças , Camundongos
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa