Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 10.710
Filtrar
2.
FASEB J ; 38(13): e23806, 2024 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-38970404

RESUMO

Atherosclerosis refers to a disease characterized by the formation of lipid plaque deposits within arterial walls, leading to reduced blood flow or blockage of blood outflow. The process of endothelial injury induced by oxidized low-density lipoprotein (ox-LDL) is considered the initial stage of atherosclerosis. Ferroptosis is a form of iron-dependent, non-apoptotic cell death, and current research suggests its association with coronary artery disease (CAD). In this study, we observed a correlation between reduced expression of SREBP-1 and the occurrence of stable CAD. Additionally, during the process of endothelial injury induced by ox-LDL, we also noted decreased expression of the SREBP-1/SCD1/FADS2 and involvement in the ferroptosis process. Mechanistically, ox-LDL induced endothelial injury by inhibiting the lipid biosynthesis process mediated by the SREBP-1/SCD1/FADS2, thereby inducing lipid peroxidation and ferroptosis. On the contrary, overexpression of SREBP-1 or supplementation with monounsaturated fatty acids counteracted iron accumulation, mitochondrial damage, and lipid peroxidation-induced ferroptosis, thereby improving endothelial injury. Our study indicated that the decreased expression of peripheral blood SREBP-1 mRNA is an independent risk factor for stable CAD. Furthermore, in endothelial cells, the lipid biosynthesis process mediated by SREBP-1 could ameliorate endothelial injury by resisting ferroptosis. The study has been registered with the Chinese Clinical Trial Registry, which serves as a primary registry in the World Health Organization International Clinical Trials Registry Platform (ChiCTR2300074315, August 3rd, 2023).


Assuntos
Ferroptose , Lipogênese , Lipoproteínas LDL , Proteína de Ligação a Elemento Regulador de Esterol 1 , Humanos , Proteína de Ligação a Elemento Regulador de Esterol 1/metabolismo , Proteína de Ligação a Elemento Regulador de Esterol 1/genética , Masculino , Lipoproteínas LDL/metabolismo , Feminino , Peroxidação de Lipídeos , Células Endoteliais da Veia Umbilical Humana/metabolismo , Doença da Artéria Coronariana/metabolismo , Doença da Artéria Coronariana/patologia , Pessoa de Meia-Idade , Células Endoteliais/metabolismo , Aterosclerose/metabolismo , Aterosclerose/patologia , Estearoil-CoA Dessaturase/metabolismo , Estearoil-CoA Dessaturase/genética , Idoso
3.
Exp Biol Med (Maywood) ; 249: 10185, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38978540

RESUMO

Atherosclerosis has traditionally been considered as a disorder characterized by the accumulation of cholesterol and thrombotic materials within the arterial wall. However, it is now understood to be a complex inflammatory disease involving multiple factors. Central to the pathogenesis of atherosclerosis are the interactions among monocytes, macrophages, and neutrophils, which play pivotal roles in the initiation, progression, and destabilization of atherosclerotic lesions. Recent advances in our understanding of atherosclerosis pathogenesis, coupled with results obtained from experimental interventions, lead us to propose the hypothesis that atherosclerosis may be reversible. This paper outlines the evolution of this hypothesis and presents corroborating evidence that supports the potential for atherosclerosis regression through the restoration of vascular copper homeostasis. We posit that these insights may pave the way for innovative therapeutic approaches aimed at the reversal of atherosclerosis.


Assuntos
Aterosclerose , Cobre , Homeostase , Cobre/metabolismo , Aterosclerose/metabolismo , Aterosclerose/patologia , Humanos , Animais
4.
Physiology (Bethesda) ; 39(5): 0, 2024 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-38984789

RESUMO

Alterations in vascular extracellular matrix (ECM) components, interactions, and mechanical properties influence both the formation and stability of atherosclerotic plaques. This review discusses the contribution of the ECM microenvironment in vascular homeostasis and remodeling in atherosclerosis, highlighting Cartilage oligomeric matrix protein (COMP) and its degrading enzyme ADAMTS7 as examples, and proposes potential avenues for future research aimed at identifying novel therapeutic targets for atherosclerosis based on the ECM microenvironment.


Assuntos
Aterosclerose , Matriz Extracelular , Homeostase , Humanos , Aterosclerose/metabolismo , Aterosclerose/fisiopatologia , Aterosclerose/patologia , Animais , Matriz Extracelular/metabolismo , Homeostase/fisiologia , Proteína de Matriz Oligomérica de Cartilagem/metabolismo , Remodelação Vascular/fisiologia
5.
Front Immunol ; 15: 1410832, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38975335

RESUMO

Introduction: Aging increases the risk of atherosclerotic vascular disease and its complications. Macrophages are pivotal in the pathogenesis of vascular aging, driving inflammation and atherosclerosis progression. NOX4 (NADPH oxidase 4) expression increases with age, correlating with mitochondrial dysfunction, inflammation, and atherosclerosis. We hypothesized that the NOX4-dependent mitochondrial oxidative stress promotes aging-associated atherosclerosis progression by causing metabolic dysfunction and inflammatory phenotype switch in macrophages. Methods: We studied atherosclerotic lesion morphology and macrophage phenotype in young (5-month-old) and aged (16-month-old) Nox4 -/-/Apoe -/- and Apoe -/- mice fed Western diet. Results: Young Nox4-/-/Apoe-/- and Apoe-/- mice had comparable aortic and brachiocephalic artery atherosclerotic lesion cross-sectional areas. Aged mice showed significantly increased lesion area compared with young mice. Aged Nox4-/-/Apoe-/- had significantly lower lesion areas than Apoe-/- mice. Compared with Apoe-/- mice, atherosclerotic lesions in aged Nox4-/-/Apoe-/- showed reduced cellular and mitochondrial ROS and oxidative DNA damage, lower necrotic core area, higher collagen content, and decreased inflammatory cytokine expression. Immunofluorescence and flow cytometry analysis revealed that aged Apoe-/- mice had a higher percentage of classically activated pro-inflammatory macrophages (CD38+CD80+) in the lesions. Aged Nox4-/-/Apoe-/- mice had a significantly higher proportion of alternatively activated pro-resolving macrophages (EGR2+/CD163+CD206+) in the lesions, with an increased CD38+/EGR2+ cell ratio compared with Apoe-/- mice. Mitochondrial respiration assessment revealed impaired oxidative phosphorylation and increased glycolytic ATP production in macrophages from aged Apoe-/- mice. In contrast, macrophages from Nox4-/-/Apoe-/- mice were less glycolytic and more aerobic, with preserved basal and maximal respiration and mitochondrial ATP production. Macrophages from Nox4-/-/Apoe-/- mice also had lower mitochondrial ROS levels and reduced IL1ß secretion; flow cytometry analysis showed fewer CD38+ cells after IFNγ+LPS treatment and more EGR2+ cells after IL4 treatment than in Apoe-/- macrophages. In aged Apoe-/- mice, inhibition of NOX4 activity using GKT137831 significantly reduced macrophage mitochondrial ROS and improved mitochondrial function, resulting in decreased CD68+CD80+ and increased CD163+CD206+ lesion macrophage proportion and attenuated atherosclerosis. Discussion: Our findings suggest that increased NOX4 in aging drives macrophage mitochondrial dysfunction, glycolytic metabolic switch, and pro-inflammatory phenotype, advancing atherosclerosis. Inhibiting NOX4 or mitochondrial dysfunction could alleviate vascular inflammation and atherosclerosis, preserving plaque integrity.


Assuntos
Envelhecimento , Aterosclerose , Macrófagos , Mitocôndrias , NADPH Oxidase 4 , Fenótipo , Animais , Aterosclerose/metabolismo , Aterosclerose/patologia , Aterosclerose/etiologia , Aterosclerose/imunologia , Mitocôndrias/metabolismo , Macrófagos/imunologia , Macrófagos/metabolismo , Camundongos , Envelhecimento/imunologia , NADPH Oxidase 4/metabolismo , NADPH Oxidase 4/genética , Progressão da Doença , Camundongos Knockout , Estresse Oxidativo , Inflamação/imunologia , Inflamação/metabolismo , Camundongos Endogâmicos C57BL , Espécies Reativas de Oxigênio/metabolismo , Masculino , Modelos Animais de Doenças , Apolipoproteínas E/genética , Apolipoproteínas E/deficiência , Camundongos Knockout para ApoE , Reprogramação Metabólica
6.
Sci Rep ; 14(1): 15847, 2024 Jul 09.
Artigo em Inglês | MEDLINE | ID: mdl-38982274

RESUMO

Atherosclerosis is rare in internal thoracic arteries (ITA) even in patients with severe atherosclerotic coronary artery (ACA) disease. To explore cellular differences, ITA SMC from 3 distinct donors and ACA SMC from 3 distinct donors were grown to sub-confluence and growth arrested for 48 h. Proliferation and thrombospondin-1 (TSP1) production were determined using standard techniques. ITA SMC were larger, grew more slowly and survived more passages than ACA SMC. ACA SMC had a more pronounced proliferative response to 10% serum than ITA SMC. Both ACA SMC and ITA SMC proliferated in response to exogenous TSP1 (12.5 µg/ml and 25 µg/ml) and platelet derived growth factor-BB (PDGF-BB; 20 ng/ml) but TSP1- and PDGF-BB-induced proliferation were partially inhibited by anti-TSP1 antibody A4.1, microRNA-21(miR-21)-3p inhibitors and miR-21-5p inhibitors in each of the 3 ACA SMC lines, but not in any of the ITA SMC lines. PDGF-BB stimulated TSP1 production in ACA SMC but not in ITA SMC but there was no increase in TSP1 levels in conditioned media in either SMC type. In summary, there are significant differences in morphology, proliferative capacity and in responses to TSP1 and PDGF-BB in SMC derived from ITA compared to SMC derived from ACA.


Assuntos
Becaplermina , Proliferação de Células , Vasos Coronários , Miócitos de Músculo Liso , Trombospondina 1 , Becaplermina/metabolismo , Trombospondina 1/metabolismo , Trombospondina 1/genética , Humanos , Proliferação de Células/efeitos dos fármacos , Miócitos de Músculo Liso/metabolismo , Miócitos de Músculo Liso/efeitos dos fármacos , Vasos Coronários/metabolismo , Vasos Coronários/patologia , Vasos Coronários/efeitos dos fármacos , MicroRNAs/genética , MicroRNAs/metabolismo , Artéria Torácica Interna/metabolismo , Artéria Torácica Interna/efeitos dos fármacos , Doença da Artéria Coronariana/metabolismo , Doença da Artéria Coronariana/patologia , Aterosclerose/metabolismo , Aterosclerose/patologia , Células Cultivadas , Masculino
7.
Int J Mol Sci ; 25(13)2024 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-39000363

RESUMO

Foods enriched with insects can potentially prevent several health disorders, including cardiovascular diseases, by reducing inflammation and improving antioxidant status. In this study, Tenebrio molitor and Gryllus assimilis were selected to determine the effect on the development of atherosclerosis in ApoE/LDLR-/- mice. Animals were fed AIN-93G-based diets (control) with 10% Tenebrio molitor (TM) and 10% Gryllus assimilis (GA) for 8 weeks. The nutritional value as well as antioxidant activity of selected insects were determined. The lipid profile, liver enzyme activity, and the fatty acid composition of liver and adipose tissue of model mice were evaluated. Quantitative analysis of atherosclerotic lesions in the entire aorta was performed using the en face method, and for aortic roots, the cross-section method was used. The antioxidant status of the GA cricket was significantly higher compared to the TM larvae. The results showed that the area of atherosclerosis (en face method) was not significantly different between groups. Dietary GA reduced plaque formation in the aortic root; additionally, significant differences were observed in sections at 200 and 300 µm compared to other groups. Furthermore, liver enzyme ALT activity was lower in insect-fed groups compared to the control group. The finding suggests that a diet containing edible insect GA potentially prevents atherosclerotic plaque development in the aortic root, due to its high antioxidant activity.


Assuntos
Apolipoproteínas E , Aterosclerose , Receptores de LDL , Animais , Aterosclerose/patologia , Aterosclerose/metabolismo , Camundongos , Receptores de LDL/genética , Receptores de LDL/metabolismo , Apolipoproteínas E/deficiência , Apolipoproteínas E/genética , Insetos Comestíveis , Camundongos Knockout , Fígado/metabolismo , Fígado/patologia , Antioxidantes/metabolismo , Masculino , Tenebrio , Dieta , Aorta/patologia , Aorta/metabolismo , Modelos Animais de Doenças , Ração Animal , Placa Aterosclerótica/patologia , Placa Aterosclerótica/metabolismo , Gryllidae
8.
Zhongguo Zhong Yao Za Zhi ; 49(13): 3619-3626, 2024 Jul.
Artigo em Chinês | MEDLINE | ID: mdl-39041134

RESUMO

The intervention effect of astragaloside Ⅳ(AS-Ⅳ) on atherosclerosis in apolipoprotein E gene knockout(ApoE)~(-/-) mice was observed based on the nuclear factor erythroid-2-related factor 2(Nrf2)/heme oxygenase-1(HO-1)/glutathione peroxidase 4(GPX4) signaling pathway to explore the potential mechanism of AS-Ⅳ in improving ferroptosis in atherosclerotic mice. This study established an atherosclerosis mouse model by feeding them a high-fat diet. After modeling for 8 weeks, ApoE~(-/-) mice were randomly divided into the model group, AS-Ⅳ group, AS-Ⅳ+Nrf2 inhibitor(ML385) group, and ferrostatin-1(Fer-1) group. Additionally, a blank control group was also established. Corresponding drugs were administered via intraperitoneal injection, with the control group receiving an equivalent amount of normal saline injection as the model group. After the experiment, serum biochemical levels were measured using an automatic blood lipid analyzer, hematoxylin-eosin(HE) staining was used to observe morphological changes in aortic sinus tissues, colorimetric methods were used to detect levels of ferrous ion(Fe~(2+)), malondialdehyde(MDA), glutathione(GSH), and superoxide dismutase(SOD) in mouse serum, immunofluorescence was used to observe the expressions of ferritin heavy chain 1(FTH1) and ferritin light chain(FTL) proteins in the aortic sinus of mice, Western blot was used to detect the protein levels of Nrf2, HO-1, and GPX4 in mouse aortic tissues, and transmission electron microscopy was used to observe ultrastructural changes in aortic tissues. RESULTS:: showed that compared to the control group, the model group of mice had significantly increased calcification and plaque deposition areas in the aortic sinus, increased mitochondrial membrane density, decreased or disappeared mitochondrial cristae, elevated levels of total cholesterol(TC), triglycerides(TG), low-density lipoprotein cholesterol(LDL-C), Fe~(2+), and MDA, decreased levels of high-density lipoprotein cholesterol(HDL-C), SOD, and GSH, and significant inhibition of Nrf2, HO-1, GPX4 proteins, as well as iron storage proteins FTH1 and FTL expressions in the aorta. Compared to the model group, AS-Ⅳ treatment resulted in decreased serum TC, TG, LDL-C, Fe~(2+), and MDA levels, increased HDL-C, SOD, and GSH levels, increased expressions of Nrf2, HO-1, and GPX4 proteins, and iron storage proteins FTH1 and FTL, and significant improvement in aortic tissue morphology. Compared to the AS-Ⅳ group, the Nrf2 inhibitor ML385 could reverse the therapeutic effect of AS-Ⅳ on atherosclerosis mice. These findings suggest that AS-Ⅳ can inhibit ferroptosis and improve atherosclerosis in ApoE~(-/-) mice, and its mechanism of action may be related to the regulation of the Nrf2/HO-1/GPX4 signaling pathway.


Assuntos
Apolipoproteínas E , Aterosclerose , Ferroptose , Heme Oxigenase-1 , Fator 2 Relacionado a NF-E2 , Fosfolipídeo Hidroperóxido Glutationa Peroxidase , Saponinas , Triterpenos , Animais , Fator 2 Relacionado a NF-E2/metabolismo , Fator 2 Relacionado a NF-E2/genética , Aterosclerose/tratamento farmacológico , Aterosclerose/metabolismo , Aterosclerose/genética , Camundongos , Ferroptose/efeitos dos fármacos , Saponinas/farmacologia , Triterpenos/farmacologia , Apolipoproteínas E/genética , Masculino , Fosfolipídeo Hidroperóxido Glutationa Peroxidase/metabolismo , Fosfolipídeo Hidroperóxido Glutationa Peroxidase/genética , Heme Oxigenase-1/metabolismo , Heme Oxigenase-1/genética , Transdução de Sinais/efeitos dos fármacos , Camundongos Knockout , Humanos , Camundongos Endogâmicos C57BL
9.
Zhongguo Zhong Yao Za Zhi ; 49(11): 2973-2980, 2024 Jun.
Artigo em Chinês | MEDLINE | ID: mdl-39041157

RESUMO

This study aims to investigate the effect and mechanism of the EtO Ac extract of Draconis Sanguis(DSE) on improving athero sclerosis in ApoE gene knockout(ApoE~(-/-)) mice. The ApoE~(-/-) mice were randomly divided into five groups: control group, mo delgroup, positive group treated with ezetimibe of 5 mg·kg~(-1)(EG), and low(100 mg·kg~(-1)) and high dose(200 mg·kg~(-1)) groups ofDSE. xcept for the control group, all other groups were fed a high-fat diet and administered drugs for 16 successive weeks. After 16 weeks of Eadministration, the body weight, liver, and epididymal fat mass of the mice were measured; the level of blood lipid and the plaquearea of the aortic outflow tract were detected to evaluate the efficacy of DSE in vivo. In addition, in vitro cultures of human umbilical v ein endothelial cell(HUVEC) were conducted. Oxidative stress of endothelial cells was induced by oxidized low-density lipoprot ein(ox-LDL), and the effects of DSE on oxidative stress-related proteins in endothelial cells were examined. The results sho wedthat both doses of DSE significantly improved the epididymal fat mass and index of ApoE~(-/-) mice with atherosclerosis, lowered thelevels of plasma cholesterol, triglyceride, and non-high density lipoprotein cholesterol, and reduced the plaque area of the aortic ou tflow tract. totIn alvitro experiments confirmed that ox-LDL significantly increased the level of lipid peroxidation marker 4-HNE in HUVECcells, confirming that DSE improved the degree of atherosclerotic lesions in ApoE~(-/-) mice by inhibiting ox-LDL-induced oxidative stress in vascular endothelial cells.


Assuntos
Apolipoproteínas E , Aterosclerose , Camundongos Knockout , Animais , Aterosclerose/tratamento farmacológico , Aterosclerose/genética , Aterosclerose/metabolismo , Camundongos , Apolipoproteínas E/genética , Masculino , Humanos , Estresse Oxidativo/efeitos dos fármacos , Medicamentos de Ervas Chinesas/farmacologia , Medicamentos de Ervas Chinesas/administração & dosagem , Células Endoteliais da Veia Umbilical Humana/efeitos dos fármacos , Camundongos Endogâmicos C57BL
10.
J Pharm Biomed Anal ; 248: 116338, 2024 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-38971092

RESUMO

Tetrahydroxy stilbene glucoside (TSG) is a water-soluble natural product that has shown potential in treating atherosclerosis (AS). However, its underlying mechanisms remain unclear. Here, we demonstrate that an 8-week TSG treatment (100 mg/kg/d) significantly reduces atherosclerotic lesions and alleviates dyslipidemia symptoms in ApoE-/- mice. 1H nuclear magnetic resonance metabolomic analysis reveals differences in both lipid components and water-soluble metabolites in the livers of AS mice compared to control groups, and TSG treatment shifts the metabolic profiles of AS mice towards a normal state. At the transcriptional level, TSG significantly restores the expression of fatty acid metabolism-related genes (Srepb-1c, Fasn, Scd1, Gpat1, Dgat1, Pparα and Cpt1α), and regulates the expression levels of disturbed cholesterol metabolism-related genes (Srebp2, Hmgcr, Ldlr, Acat1, Acat2 and Cyp7a1) associated with lipid metabolism. Furthermore, at the cellular level, TSG remarkably polarizes aortic macrophages to their M2 phenotype. Our data demonstrate that TSG alleviates arthrosclerosis by dual-targeting to hepatic lipid metabolism and aortic M2 macrophage polarization in ApoE-/- mice, with significant implications for translational medicine and the treatment of AS using natural products.


Assuntos
Aorta , Apolipoproteínas E , Aterosclerose , Glucosídeos , Metabolismo dos Lipídeos , Fígado , Macrófagos , Estilbenos , Animais , Aterosclerose/tratamento farmacológico , Aterosclerose/metabolismo , Camundongos , Glucosídeos/farmacologia , Metabolismo dos Lipídeos/efeitos dos fármacos , Fígado/efeitos dos fármacos , Fígado/metabolismo , Aorta/efeitos dos fármacos , Aorta/metabolismo , Estilbenos/farmacologia , Apolipoproteínas E/genética , Masculino , Macrófagos/efeitos dos fármacos , Macrófagos/metabolismo , Camundongos Endogâmicos C57BL , Modelos Animais de Doenças , Camundongos Knockout
11.
J Am Heart Assoc ; 13(14): e034066, 2024 Jul 16.
Artigo em Inglês | MEDLINE | ID: mdl-38979792

RESUMO

BACKGROUND: Atherosclerosis is highly prevalent in people with chronic kidney disease (CKD), including those receiving peritoneal dialysis (PD). Although it is lifesaving, PD induces profound systemic inflammation, which may aggravate atherosclerosis. Therefore, the hypothesis is that this PD-induced inflammation aggravates atherosclerosis via immune cell activation. METHODS AND RESULTS: ApoE-/- mice were subjected to a 5/6 nephrectomy to induce CKD. Three weeks later, mice were fed a high-cholesterol diet. Half of the nephrectomized mice then received daily peritoneal infusions of 3.86% Physioneal for 67 further days (CKD+PD) until the end of the experiment, and were compared with mice without CKD. Sham operated and PD-only mice were additional controls. CKD+PD mice displayed more severe atherosclerotic disease than control mice. Plaque area increased, and plaques were more advanced with a vulnerable phenotype typified by decreased collagen content and decreased fibrous cap thickness. Increased CD3+ T-cell numbers were present in plaques and perivascular adipose tissue of CKD and CKD+PD mice. Plaques of CKD+PD mice contained more iNOS+ immune cells. Spleens of CKD+PD mice showed more CD4+ central memory, terminally differentiated type 1 T-helper (Th1), Th17, and CX3C motif chemokine receptor 1+ (CX3CR1) CD4+ T-cells with less regulatory and effector T-cells. CONCLUSIONS: PD-fluid exposure in uremic mice potentiates systemic and vascular T-cell-driven inflammation and aggravates atherosclerosis. PD polarized CD4+ T-cells toward an inflammatory Th1/Th17 phenotype, and increased CX3CR1+ CD4+ T-cells, which are associated with vascular homing in CKD-associated atherosclerosis. Targeting CD4+ T-cell activation and CX3CR1+ polarization has the potential to attenuate atherosclerosis in PD patients.


Assuntos
Aterosclerose , Modelos Animais de Doenças , Diálise Peritoneal , Insuficiência Renal Crônica , Uremia , Animais , Aterosclerose/patologia , Aterosclerose/etiologia , Aterosclerose/imunologia , Aterosclerose/metabolismo , Aterosclerose/genética , Uremia/imunologia , Uremia/metabolismo , Diálise Peritoneal/efeitos adversos , Insuficiência Renal Crônica/imunologia , Insuficiência Renal Crônica/metabolismo , Camundongos Knockout para ApoE , Camundongos , Placa Aterosclerótica , Masculino , Camundongos Endogâmicos C57BL , Apolipoproteínas E/genética , Apolipoproteínas E/deficiência , Nefrectomia
12.
Xi Bao Yu Fen Zi Mian Yi Xue Za Zhi ; 40(6): 527-531, 2024 Jun.
Artigo em Chinês | MEDLINE | ID: mdl-38952092

RESUMO

Objective To explore a simple and feasible method for whole-mount immunofluorescence staining of lymphatic vessels in the ApoE-/- mouse model of atherosclerosis. Methods Aortic specimens were carefully excised from the ApoE-/- mouse model. Following immunostaining with specific antibodies against smooth muscle actin (SMA) and lymphatic vessel endothelial receptor 1 (LYVE1), the aortas, including the aortic root, were subjected to a 30-minute treatment with 5 g/L Sudan Black B solution. This step was instrumental in minimizing the autofluorescent background of the tissue. Thereafter, the aortas were processed through a clearing protocol and imaged within a purpose-built chamber under a fluorescence microscope. Results The pretreatment with 5 g/L Sudan Black B effectively suppressed the autofluorescent signals emanating from the vascular structures, thereby enhancing the contrast and clarity of the specific fluorescence signals associated with the lymphatic vessels. This enhancement in signal quality did not compromise the integrity or specificity of the immunofluorescent markers. Conclusion A facile, highly specific, and effective approach for the visualization of lymphatic vessels in whole-mount aortic preparations from ApoE-/- mice is established.


Assuntos
Aorta , Apolipoproteínas E , Imunofluorescência , Vasos Linfáticos , Animais , Vasos Linfáticos/metabolismo , Vasos Linfáticos/diagnóstico por imagem , Camundongos , Aorta/metabolismo , Apolipoproteínas E/genética , Apolipoproteínas E/deficiência , Apolipoproteínas E/metabolismo , Imunofluorescência/métodos , Túnica Adventícia/metabolismo , Aterosclerose/metabolismo , Aterosclerose/patologia , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Coloração e Rotulagem/métodos , Microscopia de Fluorescência/métodos
13.
Pharmacol Res ; 206: 107289, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-38960011

RESUMO

Atherosclerosis is a chronic inflammatory vascular disease characterized by lipid metabolism disorder and lipid accumulation. Equisetin (EQST) is a hemiterpene compound isolated from fungus of marine sponge origin, which has antibacterial, anti-inflammatory, lipid-lowering, and weight loss effects. Whether EQST has anti-atherosclerotic activity has not been reported. In this study, we revealed that EQST displayed anti- atherosclerosis effects through inhibiting macrophage inflammatory response, lipid uptake and foam cell formation in vitro, and finally ameliorated high-fat diet (HFD)-induced atherosclerosis in AopE-/- mice in vivo. Mechanistically, EQST directly bound to STAT3 with high-affinity by forming hydrophobic bonds at GLN247 and GLN326 residues, as well as hydrogen bonds at ARG325 and THR346 residues. EQST interacted with STAT3 physically, and functionally inhibited the transcription activity of STAT3, thereby regulating atherosclerosis. Therefore, these results supports EQST as a candidate for developing anti-atherosclerosis therapeutic agent.


Assuntos
Aterosclerose , Camundongos Endogâmicos C57BL , Fator de Transcrição STAT3 , Fator de Transcrição STAT3/metabolismo , Animais , Aterosclerose/tratamento farmacológico , Aterosclerose/metabolismo , Aterosclerose/prevenção & controle , Camundongos , Masculino , Dieta Hiperlipídica/efeitos adversos , Humanos , Células RAW 264.7 , Camundongos Knockout , Ligação Proteica , Anti-Inflamatórios/farmacologia , Anti-Inflamatórios/uso terapêutico , Células Espumosas/efeitos dos fármacos , Células Espumosas/metabolismo
14.
Clinics (Sao Paulo) ; 79: 100413, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-39024795

RESUMO

OBJECTIVE: Atherosclerosis (AS) is a common pathogenesis of cardiovascular diseases. Puerarin (Pue) is a Chinese herbal remedy used to prevent and treat AS. Here, this research investigated the effect of Pue on AS progression. METHODS: ApoE-/- mice were induced with acrolein. Body weight, blood lipid index, inflammatory factors, mitochondrial oxidative stress, and lipid deposition were detected. IL-6 and TNF-α were detected by ELISA. Oil red staining and H&E staining were used to observe the aortic sinus plaque lesions. Serum expressions of inflammatory factors IL-6, TNF-a, SOD, GSH and MDA were detected by ELISA, the mRNA expression levels of HDAC1 in the aorta were detected by RT-qPCR, and IL-6 and TNF-α in the aorta were detected by immunohistochemistry. JNK, p-JNK, OPA-1, and HDAC1 were detected by Western blotting. RESULTS: Pue administration can effectively reduce lipid accumulation in AS mice induced by acrolein. Pue promoted the activity of SOD, GSH and MDA, and inhibited the formation of atherosclerotic plaques and the process of aortic histological changes. Pue reduced IL-6 and TNF-α. HDAC1 expression was down-regulated and p-JNK-1 and JNK protein expression was up-regulated. CONCLUSION: Pue reduces inflammation and alleviates AS induced by acrolein by mediating the JNK pathway to inhibit HDAC1-mediated oxidative stress disorder.


Assuntos
Acroleína , Aterosclerose , Histona Desacetilase 1 , Isoflavonas , Estresse Oxidativo , Animais , Aterosclerose/induzido quimicamente , Aterosclerose/metabolismo , Aterosclerose/tratamento farmacológico , Estresse Oxidativo/efeitos dos fármacos , Histona Desacetilase 1/metabolismo , Isoflavonas/farmacologia , Isoflavonas/uso terapêutico , Acroleína/farmacologia , Masculino , Fator de Necrose Tumoral alfa/metabolismo , Interleucina-6/metabolismo , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Camundongos , Modelos Animais de Doenças , Ensaio de Imunoadsorção Enzimática , Western Blotting , Aorta/efeitos dos fármacos , Aorta/patologia
16.
Sci Rep ; 14(1): 16364, 2024 Jul 16.
Artigo em Inglês | MEDLINE | ID: mdl-39013959

RESUMO

Non-alcoholic steatohepatitis (NASH) is a hepatocyte inflammation based on hepatocellular steatosis, yet there is no effective drug treatment. Atherosclerosis (AS) is caused by lipid deposition in the endothelium, which can lead to various cardiovascular diseases. NASH and AS share common risk factors, and NASH can also elevate the risk of AS, causing a higher morbidity and mortality rate for atherosclerotic heart disease. Therefore, timely detection and diagnosis of NASH and AS are particularly important. In this study, differential gene expression analysis and weighted gene co-expression network analysis were performed on the AS (GSE100927) and NASH (GSE89632) datasets to obtain common crosstalk genes, respectively. Then, candidate Hub genes were screened using four topological algorithms and externally validated in the GSE43292 and GSE63067 datasets to obtain Hub genes. Furthermore, immune infiltration analysis and gene set variation analysis were performed on the Hub genes to explore the underlying mechanisms. The DGIbd database was used to screen candidate drugs for AS and NASH. Finally, a NASH model was constructed using free fatty acid-induced human L02 cells, an AS model was constructed using lipopolysaccharide-induced HUVECs, and a co-morbidity model was constructed using L02 cells and HUVECs to verify Hub gene expression. The result showed that a total of 113 genes common to both AS and NASH were identified as crosstalk genes, and enrichment analysis indicated that these genes were mainly involved in the regulation of immune and metabolism-related pathways. 28 candidate Hub genes were screened according to four topological algorithms, and CXCL9, IL2RB, and SPP1 were identified as Hub genes after in vitro experiments and external dataset validation. The ROC curves and SVM modeling demonstrated the good diagnostic efficacy of these three Hub genes. In addition, the Hub genes are strongly associated with immune cell infiltration, especially macrophages and γ-δ T cell infiltration. Finally, five potential therapeutic drugs were identified. has-miR-185 and hsa-miR-335 were closely related to AS and NASH. This study demonstrates that CXCL9, IL2RB, and SPP1 may serve as potential biomarkers for the diagnosis of the co-morbidity patterns of AS and NASH and as potential targets for drug therapy.


Assuntos
Aterosclerose , Biomarcadores , Quimiocina CXCL9 , Hepatopatia Gordurosa não Alcoólica , Humanos , Hepatopatia Gordurosa não Alcoólica/genética , Hepatopatia Gordurosa não Alcoólica/diagnóstico , Hepatopatia Gordurosa não Alcoólica/metabolismo , Hepatopatia Gordurosa não Alcoólica/epidemiologia , Hepatopatia Gordurosa não Alcoólica/patologia , Aterosclerose/genética , Aterosclerose/metabolismo , Aterosclerose/diagnóstico , Biomarcadores/metabolismo , Quimiocina CXCL9/genética , Quimiocina CXCL9/metabolismo , Redes Reguladoras de Genes , Comorbidade , Células Endoteliais da Veia Umbilical Humana/metabolismo , Perfilação da Expressão Gênica
17.
Medicine (Baltimore) ; 103(29): e38896, 2024 Jul 19.
Artigo em Inglês | MEDLINE | ID: mdl-39029045

RESUMO

BACKGROUND: The purpose of this study was to investigate the role of legumain in metabolic dysfunction, diagnosis, and prognosis in patients with atherosclerosis. METHODS: Plasma levels of legumain from patients with atherosclerosis (n = 320) and healthy controls (n = 320), expression of legumain in atheromatous plaque and secreted from monocyte-derived macrophages were measured using enzyme-linked-immunosorbent assay, reverse transcription-polymerase chain reaction, Western blot, immunohistochemistry, and fluorescence. RESULTS: Data demonstrated that atherosclerotic patients had higher plasma level of legumain than healthy controls, which was a diagnostic and prognostic marker and corrected with the degree of atherosclerosis. It found that atheromatous plaque and endothelial cell had higher legumain expression than non-atherosclerotic arteries (controls). Legumain showed significantly increased secretion from pro-inflammatory M1 compared to pro-resolving M2 macrophages during monocyte-derived macrophages, which was localized to structures resembling foam cells. CONCLUSION: In conclusion, our data indicate that legumain expression is upregulated in both plasma and plaques of patients with atherosclerosis, which is associated with metabolic dysfunction of endothelial cell and might be a diagnostic and prognostic marker of atherosclerosis.


Assuntos
Aterosclerose , Biomarcadores , Cisteína Endopeptidases , Macrófagos , Humanos , Cisteína Endopeptidases/metabolismo , Cisteína Endopeptidases/sangue , Masculino , Feminino , Aterosclerose/diagnóstico , Aterosclerose/metabolismo , Prognóstico , Pessoa de Meia-Idade , Macrófagos/metabolismo , Biomarcadores/sangue , Biomarcadores/metabolismo , Placa Aterosclerótica/metabolismo , Idoso , Estudos de Casos e Controles , Regulação para Cima , Adulto
18.
Bull Math Biol ; 86(8): 104, 2024 Jul 09.
Artigo em Inglês | MEDLINE | ID: mdl-38980556

RESUMO

Atherosclerotic plaques are fatty deposits that form in the walls of major arteries and are one of the major causes of heart attacks and strokes. Macrophages are the main immune cells in plaques and macrophage dynamics influence whether plaques grow or regress. Macrophage proliferation is a key process in atherosclerosis, particularly in the development of mid-stage plaques, but very few mathematical models include proliferation. In this paper we reframe the lipid-structured model of Ford et al. (J Theor Biol 479:48-63, 2019. https://doi.org/10.1016/j.jtbi.2019.07.003 ) to account for macrophage proliferation. Proliferation is modelled as a non-local decrease in the lipid structural variable. Steady state analysis indicates that proliferation assists in reducing eventual necrotic core lipid content and spreads the lipid load of the macrophage population amongst the cells. The contribution of plaque macrophages from proliferation relative to recruitment from the bloodstream is also examined. The model suggests that a more proliferative plaque differs from an equivalent (defined as having the same lipid content and cell numbers) recruitment-dominant plaque in the way lipid is distributed amongst the macrophages. The macrophage lipid distribution of an equivalent proliferation-dominant plaque is less skewed and exhibits a local maximum near the endogenous lipid content.


Assuntos
Aterosclerose , Proliferação de Células , Metabolismo dos Lipídeos , Macrófagos , Conceitos Matemáticos , Modelos Cardiovasculares , Placa Aterosclerótica , Macrófagos/patologia , Macrófagos/metabolismo , Aterosclerose/patologia , Aterosclerose/metabolismo , Placa Aterosclerótica/patologia , Humanos , Animais , Simulação por Computador , Lipídeos
19.
J Cell Mol Med ; 28(10): e18402, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-39008328

RESUMO

Syntaxin 17 (STX17) has been identified as a crucial factor in mediating the fusion of autophagosomes and lysosomes. However, its specific involvement in the context of atherosclerosis (AS) remains unclear. This study sought to elucidate the role and mechanistic contributions of STX17 in the initiation and progression of AS. Utilizing both in vivo and in vitro AS model systems, we employed ApoE knockout (KO) mice subjected to a high-fat diet and human umbilical vein endothelial cells (HUVECs) treated with oxidized low-density lipoprotein (ox-LDL) to assess STX17 expression. To investigate underlying mechanisms, we employed shRNA-STX17 lentivirus to knock down STX17 expression, followed by evaluating autophagy and inflammation in HUVECs. In both in vivo and in vitro AS models, STX17 expression was significantly upregulated. Knockdown of STX17 exacerbated HUVEC damage, both with and without ox-LDL treatment. Additionally, we observed that STX17 knockdown impaired autophagosome degradation, impeded autophagy flux and also resulted in the accumulation of dysfunctional lysosomes in HUVECs. Moreover, STX17 knockdown intensified the inflammatory response following ox-LDL treatment in HUVECs. Further mechanistic exploration revealed an association between STX17 and STING; reducing STX17 expression increased STING levels. Further knockdown of STING enhanced autophagy flux. In summary, our findings suggest that STX17 knockdown worsens AS by impeding autophagy flux and amplifying the inflammatory response. Additionally, the interaction between STX17 and STING may play a crucial role in STX17-mediated autophagy.


Assuntos
Aterosclerose , Autofagia , Células Endoteliais da Veia Umbilical Humana , Inflamação , Lipoproteínas LDL , Proteínas Qa-SNARE , Autofagia/genética , Animais , Humanos , Aterosclerose/metabolismo , Aterosclerose/genética , Aterosclerose/patologia , Células Endoteliais da Veia Umbilical Humana/metabolismo , Inflamação/metabolismo , Inflamação/patologia , Inflamação/genética , Proteínas Qa-SNARE/metabolismo , Proteínas Qa-SNARE/genética , Camundongos , Lipoproteínas LDL/metabolismo , Técnicas de Silenciamento de Genes , Lisossomos/metabolismo , Camundongos Knockout , Masculino , Camundongos Endogâmicos C57BL , Modelos Animais de Doenças , Dieta Hiperlipídica/efeitos adversos , Apolipoproteínas E/genética , Apolipoproteínas E/metabolismo , Apolipoproteínas E/deficiência
20.
Nat Commun ; 15(1): 4772, 2024 Jun 10.
Artigo em Inglês | MEDLINE | ID: mdl-38858384

RESUMO

The underlying mechanisms of atherosclerosis, the second leading cause of death among Werner syndrome (WS) patients, are not fully understood. Here, we establish an in vitro co-culture system using macrophages (iMφs), vascular endothelial cells (iVECs), and vascular smooth muscle cells (iVSMCs) derived from induced pluripotent stem cells. In co-culture, WS-iMφs induces endothelial dysfunction in WS-iVECs and characteristics of the synthetic phenotype in WS-iVSMCs. Transcriptomics and open chromatin analysis reveal accelerated activation of type I interferon signaling and reduced chromatin accessibility of several transcriptional binding sites required for cellular homeostasis in WS-iMφs. Furthermore, the H3K9me3 levels show an inverse correlation with retrotransposable elements, and retrotransposable element-derived double-stranded RNA activates the DExH-box helicase 58 (DHX58)-dependent cytoplasmic RNA sensing pathway in WS-iMφs. Conversely, silencing type I interferon signaling in WS-iMφs rescues cell proliferation and suppresses cellular senescence and inflammation. These findings suggest that Mφ-specific inhibition of type I interferon signaling could be targeted to treat atherosclerosis in WS patients.


Assuntos
Aterosclerose , Inflamação , Interferon Tipo I , Macrófagos , Retroelementos , Síndrome de Werner , Interferon Tipo I/metabolismo , Síndrome de Werner/genética , Síndrome de Werner/metabolismo , Humanos , Aterosclerose/metabolismo , Aterosclerose/imunologia , Aterosclerose/genética , Aterosclerose/patologia , Macrófagos/metabolismo , Macrófagos/imunologia , Retroelementos/genética , Inflamação/metabolismo , Inflamação/patologia , Inflamação/genética , Células-Tronco Pluripotentes Induzidas/metabolismo , Transdução de Sinais , Técnicas de Cocultura , Miócitos de Músculo Liso/metabolismo , Células Endoteliais/metabolismo , Músculo Liso Vascular/metabolismo , Músculo Liso Vascular/patologia , RNA Helicases DEAD-box/metabolismo , RNA Helicases DEAD-box/genética , Senescência Celular , Proliferação de Células
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA