Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 3.391
Filtrer
1.
Int J Mol Sci ; 25(13)2024 Jul 01.
Article de Anglais | MEDLINE | ID: mdl-39000363

RÉSUMÉ

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.


Sujet(s)
Apolipoprotéines E , Athérosclérose , Récepteurs aux lipoprotéines LDL , Animaux , Athérosclérose/anatomopathologie , Athérosclérose/métabolisme , Souris , Récepteurs aux lipoprotéines LDL/génétique , Récepteurs aux lipoprotéines LDL/métabolisme , Apolipoprotéines E/déficit , Apolipoprotéines E/génétique , Insectes comestibles , Souris knockout , Foie/métabolisme , Foie/anatomopathologie , Antioxydants/métabolisme , Mâle , Tenebrio , Régime alimentaire , Aorte/anatomopathologie , Aorte/métabolisme , Modèles animaux de maladie humaine , Aliment pour animaux , Plaque d'athérosclérose/anatomopathologie , Plaque d'athérosclérose/métabolisme , Gryllidae
2.
J Cell Mol Med ; 28(10): e18402, 2024 May.
Article de Anglais | MEDLINE | ID: mdl-39008328

RÉSUMÉ

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.


Sujet(s)
Athérosclérose , Autophagie , Cellules endothéliales de la veine ombilicale humaine , Inflammation , Lipoprotéines LDL , Protéines Qa-SNARE , Autophagie/génétique , Animaux , Humains , Athérosclérose/métabolisme , Athérosclérose/génétique , Athérosclérose/anatomopathologie , Cellules endothéliales de la veine ombilicale humaine/métabolisme , Inflammation/métabolisme , Inflammation/anatomopathologie , Inflammation/génétique , Protéines Qa-SNARE/métabolisme , Protéines Qa-SNARE/génétique , Souris , Lipoprotéines LDL/métabolisme , Techniques de knock-down de gènes , Lysosomes/métabolisme , Souris knockout , Mâle , Souris de lignée C57BL , Modèles animaux de maladie humaine , Alimentation riche en graisse/effets indésirables , Apolipoprotéines E/génétique , Apolipoprotéines E/métabolisme , Apolipoprotéines E/déficit
3.
Xi Bao Yu Fen Zi Mian Yi Xue Za Zhi ; 40(6): 527-531, 2024 Jun.
Article de Chinois | MEDLINE | ID: mdl-38952092

RÉSUMÉ

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.


Sujet(s)
Aorte , Apolipoprotéines E , Technique d'immunofluorescence , Vaisseaux lymphatiques , Animaux , Vaisseaux lymphatiques/métabolisme , Vaisseaux lymphatiques/imagerie diagnostique , Souris , Aorte/métabolisme , Apolipoprotéines E/génétique , Apolipoprotéines E/déficit , Apolipoprotéines E/métabolisme , Technique d'immunofluorescence/méthodes , Adventice/métabolisme , Athérosclérose/métabolisme , Athérosclérose/anatomopathologie , Mâle , Souris de lignée C57BL , Souris knockout , Coloration et marquage/méthodes , Microscopie de fluorescence/méthodes
4.
J Pineal Res ; 76(5): e12988, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-38982751

RÉSUMÉ

Vulnerable atherosclerotic plaque rupture, the leading cause of fatal atherothrombotic events, is associated with an increased risk of mortality worldwide. Peroxisome proliferator-activated receptor delta (PPARδ) has been shown to modulate vascular smooth muscle cell (SMC) phenotypic switching, and, hence, atherosclerotic plaque stability. Melatonin reportedly plays a beneficial role in cardiovascular diseases; however, the mechanisms underlying improvements in atherosclerotic plaque vulnerability remain unknown. In this study, we assessed the role of melatonin in regulating SMC phenotypic switching and its consequential contribution to the amelioration of atherosclerotic plaque vulnerability and explored the mechanisms underlying this process. We analyzed features of atherosclerotic plaque vulnerability and markers of SMC phenotypic transition in high-cholesterol diet (HCD)-fed apolipoprotein E knockout (ApoE-/-) mice and human aortic SMCs (HASMCs). Melatonin reduced atherosclerotic plaque size and necrotic core area while enhancing collagen content, fibrous cap thickness, and smooth muscle alpha-actin positive cell coverage on the plaque cap, which are all known phenotypic characteristics of vulnerable plaques. In atherosclerotic lesions, melatonin significantly decreased the synthetic SMC phenotype and KLF4 expression and increased the expression of PPARδ, but not PPARα and PPARγ, in HCD-fed ApoE-/- mice. These results were subsequently confirmed in the melatonin-treated HASMCs. Further analysis using PPARδ silencing and immunoprecipitation assays revealed that PPARδ plays a role in the melatonin-induced SMC phenotype switching from synthetic to contractile. Collectively, we provided the first evidence that melatonin mediates its protective effect against plaque destabilization by enhancing PPARδ-mediated SMC phenotypic switching, thereby indicating the potential of melatonin in treating atherosclerosis.


Sujet(s)
Facteur-4 de type Kruppel , Mélatonine , Myocytes du muscle lisse , Récepteur PPAR delta , Plaque d'athérosclérose , Animaux , Mélatonine/pharmacologie , Plaque d'athérosclérose/métabolisme , Plaque d'athérosclérose/anatomopathologie , Souris , Myocytes du muscle lisse/métabolisme , Myocytes du muscle lisse/effets des médicaments et des substances chimiques , Myocytes du muscle lisse/anatomopathologie , Facteur-4 de type Kruppel/métabolisme , Humains , Récepteur PPAR delta/métabolisme , Récepteur PPAR delta/génétique , Souris knockout , Mâle , Souris invalidées pour les gènes ApoE , Phénotype , Apolipoprotéines E/génétique , Apolipoprotéines E/métabolisme , Apolipoprotéines E/déficit , Muscles lisses vasculaires/métabolisme , Muscles lisses vasculaires/anatomopathologie , Muscles lisses vasculaires/effets des médicaments et des substances chimiques , Souris de lignée C57BL
5.
Front Immunol ; 15: 1410832, 2024.
Article de Anglais | MEDLINE | ID: mdl-38975335

RÉSUMÉ

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.


Sujet(s)
Vieillissement , Athérosclérose , Macrophages , Mitochondries , NADPH Oxidase 4 , Phénotype , Animaux , Athérosclérose/métabolisme , Athérosclérose/anatomopathologie , Athérosclérose/étiologie , Athérosclérose/immunologie , Mitochondries/métabolisme , Macrophages/immunologie , Macrophages/métabolisme , Souris , Vieillissement/immunologie , NADPH Oxidase 4/métabolisme , NADPH Oxidase 4/génétique , Évolution de la maladie , Souris knockout , Stress oxydatif , Inflammation/immunologie , Inflammation/métabolisme , Souris de lignée C57BL , Espèces réactives de l'oxygène/métabolisme , Mâle , Modèles animaux de maladie humaine , Apolipoprotéines E/génétique , Apolipoprotéines E/déficit , Souris invalidées pour les gènes ApoE ,
6.
J Am Heart Assoc ; 13(14): e034066, 2024 Jul 16.
Article de Anglais | MEDLINE | ID: mdl-38979792

RÉSUMÉ

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.


Sujet(s)
Athérosclérose , Modèles animaux de maladie humaine , Dialyse péritonéale , Insuffisance rénale chronique , Urémie , Animaux , Athérosclérose/anatomopathologie , Athérosclérose/étiologie , Athérosclérose/immunologie , Athérosclérose/métabolisme , Athérosclérose/génétique , Urémie/immunologie , Urémie/métabolisme , Dialyse péritonéale/effets indésirables , Insuffisance rénale chronique/immunologie , Insuffisance rénale chronique/métabolisme , Souris invalidées pour les gènes ApoE , Souris , Plaque d'athérosclérose , Mâle , Souris de lignée C57BL , Apolipoprotéines E/génétique , Apolipoprotéines E/déficit , Néphrectomie
7.
Sci Rep ; 14(1): 12917, 2024 06 05.
Article de Anglais | MEDLINE | ID: mdl-38839811

RÉSUMÉ

Allii Macrostemonis Bulbus (AMB) is a traditional Chinese medicine with medicinal and food homology. AMB has various biological activities, including anti-coagulation, lipid-lowering, anti-tumor, and antioxidant effects. Saponins from Allium macrostemonis Bulbus (SAMB), the predominant beneficial compounds, also exhibited lipid-lowering and anti-inflammatory properties. However, the effect of SAMB on atherosclerosis and the underlying mechanisms are still unclear. This study aimed to elucidate the pharmacological impact of SAMB on atherosclerosis. In apolipoprotein E deficiency (ApoE-/-) mice with high-fat diet feeding, oral SAMB administration significantly attenuated inflammation and atherosclerosis plaque formation. The in vitro experiments demonstrated that SAMB effectively suppressed oxidized-LDL-induced foam cell formation by down-regulating CD36 expression, thereby inhibiting lipid endocytosis in bone marrow-derived macrophages. Additionally, SAMB effectively blocked LPS-induced inflammatory response in bone marrow-derived macrophages potentially through modulating the NF-κB/NLRP3 pathway. In conclusion, SAMB exhibits a potential anti-atherosclerotic effect by inhibiting macrophage foam cell formation and inflammation. These findings provide novel insights into potential preventive and therapeutic strategies for the clinical management of atherosclerosis.


Sujet(s)
Athérosclérose , Cellules spumeuses , Inflammation , Saponines , Animaux , Cellules spumeuses/effets des médicaments et des substances chimiques , Cellules spumeuses/métabolisme , Athérosclérose/traitement médicamenteux , Athérosclérose/anatomopathologie , Athérosclérose/métabolisme , Athérosclérose/prévention et contrôle , Saponines/pharmacologie , Souris , Inflammation/traitement médicamenteux , Inflammation/anatomopathologie , Allium/composition chimique , Mâle , Apolipoprotéines E/déficit , Alimentation riche en graisse/effets indésirables , Facteur de transcription NF-kappa B/métabolisme , Souris de lignée C57BL , Lipoprotéines LDL/métabolisme
8.
Int J Mol Sci ; 25(11)2024 May 27.
Article de Anglais | MEDLINE | ID: mdl-38891996

RÉSUMÉ

Human abdominal aortic aneurysms (AAAs) are characterized by increased activity of matrix metalloproteinases (MMP), including MMP-12, alongside macrophage accumulation and elastin degradation, in conjunction with superimposed atherosclerosis. Previous genetic ablation studies have proposed contradictory roles for MMP-12 in AAA development. In this study, we aimed to elucidate if pharmacological inhibition of MMP-12 activity with a phosphinic peptide inhibitor protects from AAA formation and progression in angiotensin (Ang) II-infused Apoe-/- mice. Complimentary studies were conducted in a human ex vivo model of early aneurysm development. Administration of an MMP-12 inhibitor (RXP470.1) protected hypercholesterolemia Apoe-/- mice from Ang II-induced AAA formation and rupture-related death, associated with diminished medial thinning and elastin fragmentation alongside increased collagen deposition. Proteomic analyses confirmed a beneficial effect of MMP-12 inhibition on extracellular matrix remodeling proteins combined with inflammatory pathways. Furthermore, RXP470.1 treatment of mice with pre-existing AAAs exerted beneficial effects as observed through suppressed aortic dilation and rupture, medial thinning, and elastin destruction. Our findings indicate that pharmacological inhibition of MMP-12 activity retards AAA progression and improves survival in mice providing proof-of-concept evidence to motivate translational work for MMP-12 inhibitor therapy in humans.


Sujet(s)
Angiotensine-II , Anévrysme de l'aorte abdominale , Apolipoprotéines E , Matrix metalloproteinase 12 , Inhibiteurs de métalloprotéinases matricielles , Animaux , Anévrysme de l'aorte abdominale/métabolisme , Anévrysme de l'aorte abdominale/anatomopathologie , Anévrysme de l'aorte abdominale/induit chimiquement , Anévrysme de l'aorte abdominale/prévention et contrôle , Anévrysme de l'aorte abdominale/étiologie , Matrix metalloproteinase 12/métabolisme , Souris , Apolipoprotéines E/déficit , Apolipoprotéines E/génétique , Humains , Inhibiteurs de métalloprotéinases matricielles/pharmacologie , Inhibiteurs de métalloprotéinases matricielles/usage thérapeutique , Mâle , Modèles animaux de maladie humaine , Souris knockout , Souris de lignée C57BL , Élastine/métabolisme , Protéomique/méthodes
9.
Biomolecules ; 14(6)2024 Jun 19.
Article de Anglais | MEDLINE | ID: mdl-38927129

RÉSUMÉ

Abdominal aortic aneurysm (AAA) is a chronic aortic disease that lacks effective pharmacological therapies. This study was performed to determine the influence of treatment with the gasdermin D inhibitor necrosulfonamide on experimental AAAs. AAAs were induced in male apolipoprotein E-deficient mice by subcutaneous angiotensin II infusion (1000 ng/kg body weight/min), with daily administration of necrosulfonamide (5 mg/kg body weight) or vehicle starting 3 days prior to angiotensin II infusion for 30 days. Necrosulfonamide treatment remarkably suppressed AAA enlargement, as indicated by reduced suprarenal maximal external diameter and surface area, and lowered the incidence and reduced the severity of experimental AAAs. Histologically, necrosulfonamide treatment attenuated medial elastin breaks, smooth muscle cell depletion, and aortic wall collagen deposition. Macrophages, CD4+ T cells, CD8+ T cells, and neovessels were reduced in the aneurysmal aortas of necrosulfonamide- as compared to vehicle-treated angiotensin II-infused mice. Atherosclerosis and intimal macrophages were also substantially reduced in suprarenal aortas from angiotensin II-infused mice following necrosulfonamide treatment. Additionally, the levels of serum interleukin-1ß and interleukin-18 were significantly lower in necrosulfonamide- than in vehicle-treated mice without affecting body weight gain, lipid levels, or blood pressure. Our findings indicate that necrosulfonamide reduced experimental AAAs by preserving aortic structural integrity as well as reducing mural leukocyte accumulation, neovessel formation, and systemic levels of interleukin-1ß and interleukin-18. Thus, pharmacologically inhibiting gasdermin D activity may lead to the establishment of nonsurgical therapies for clinical AAA disease.


Sujet(s)
Angiotensine-II , Anévrysme de l'aorte abdominale , Apolipoprotéines E , Sulfonamides , Animaux , Anévrysme de l'aorte abdominale/induit chimiquement , Anévrysme de l'aorte abdominale/traitement médicamenteux , Anévrysme de l'aorte abdominale/métabolisme , Anévrysme de l'aorte abdominale/anatomopathologie , Anévrysme de l'aorte abdominale/prévention et contrôle , Souris , Mâle , Sulfonamides/pharmacologie , Apolipoprotéines E/déficit , Apolipoprotéines E/génétique , Protéines de liaison aux phosphates/métabolisme , Modèles animaux de maladie humaine , Souris de lignée C57BL , Macrophages/effets des médicaments et des substances chimiques , Macrophages/métabolisme , Indoles/pharmacologie , Souris invalidées pour les gènes ApoE , Gasdermines
10.
Redox Biol ; 74: 103229, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-38870781

RÉSUMÉ

BACKGROUND: Nuclear erythroid 2-related factor 2 (Nrf2), a transcription factor, is critically involved in the regulation of oxidative stress and inflammation. However, the role of endothelial Nrf2 in atherogenesis has yet to be defined. In addition, how endothelial Nrf2 is activated and whether Nrf2 can be targeted for the prevention and treatment of atherosclerosis is not explored. METHODS: RNA-sequencing and single-cell RNA sequencing analysis of mouse atherosclerotic aortas were used to identify the differentially expressed genes. In vivo endothelial cell (EC)-specific activation of Nrf2 was achieved by injecting adeno-associated viruses into ApoE-/- mice, while EC-specific knockdown of Nrf2 was generated in Cdh5CreCas9floxed-stopApoE-/- mice. RESULTS: Endothelial inflammation appeared as early as on day 3 after feeding of a high cholesterol diet (HCD) in ApoE-/- mice, as reflected by mRNA levels, immunostaining and global mRNA profiling, while the immunosignal of the end-product of lipid peroxidation (LPO), 4-hydroxynonenal (4-HNE), started to increase on day 10. TNF-α, 4-HNE, and erastin (LPO inducer), activated Nrf2 signaling in human ECs by increasing the mRNA and protein expression of Nrf2 target genes. Knockdown of endothelial Nrf2 resulted in augmented endothelial inflammation and LPO, and accelerated atherosclerosis in Cdh5CreCas9floxed-stopApoE-/- mice. By contrast, both EC-specific and pharmacological activation of Nrf2 inhibited endothelial inflammation, LPO, and atherogenesis. CONCLUSIONS: Upon HCD feeding in ApoE-/- mice, endothelial inflammation is an earliest event, followed by the appearance of LPO. EC-specific activation of Nrf2 inhibits atherosclerosis while EC-specific knockdown of Nrf2 results in the opposite effect. Pharmacological activators of endothelial Nrf2 may represent a novel therapeutic strategy for the treatment of atherosclerosis.


Sujet(s)
Apolipoprotéines E , Athérosclérose , Cellules endothéliales , Inflammation , Peroxydation lipidique , Facteur-2 apparenté à NF-E2 , Animaux , Humains , Mâle , Souris , Apolipoprotéines E/génétique , Apolipoprotéines E/déficit , Apolipoprotéines E/métabolisme , Athérosclérose/métabolisme , Athérosclérose/génétique , Athérosclérose/anatomopathologie , Modèles animaux de maladie humaine , Cellules endothéliales/métabolisme , Inflammation/métabolisme , Inflammation/génétique , Souris knockout , Facteur-2 apparenté à NF-E2/métabolisme , Facteur-2 apparenté à NF-E2/génétique , Stress oxydatif
11.
Int J Mol Sci ; 25(11)2024 Jun 01.
Article de Anglais | MEDLINE | ID: mdl-38892314

RÉSUMÉ

GV1001, an anticancer vaccine, exhibits other biological functions, including anti-inflammatory and antioxidant activity. It also suppresses the development of ligature-induced periodontitis in mice. Porphyromonas gingivalis (Pg), a major human oral bacterium implicated in the development of periodontitis, is associated with various systemic disorders, such as atherosclerosis and Alzheimer's disease (AD). This study aimed to explore the protective effects of GV1001 against Pg-induced periodontal disease, atherosclerosis, and AD-like conditions in Apolipoprotein (ApoE)-deficient mice. GV1001 effectively mitigated the development of Pg-induced periodontal disease, atherosclerosis, and AD-like conditions by counteracting Pg-induced local and systemic inflammation, partly by inhibiting the accumulation of Pg DNA aggregates, Pg lipopolysaccharides (LPS), and gingipains in the gingival tissue, arterial wall, and brain. GV1001 attenuated the development of atherosclerosis by inhibiting vascular inflammation, lipid deposition in the arterial wall, endothelial to mesenchymal cell transition (EndMT), the expression of Cluster of Differentiation 47 (CD47) from arterial smooth muscle cells, and the formation of foam cells in mice with Pg-induced periodontal disease. GV1001 also suppressed the accumulation of AD biomarkers in the brains of mice with periodontal disease. Overall, these findings suggest that GV1001 holds promise as a preventive agent in the development of atherosclerosis and AD-like conditions associated with periodontal disease.


Sujet(s)
Apolipoprotéines E , Athérosclérose , Maladies parodontales , Porphyromonas gingivalis , Animaux , Souris , Apolipoprotéines E/déficit , Maladies parodontales/microbiologie , Maladies parodontales/prévention et contrôle , Athérosclérose/métabolisme , Athérosclérose/prévention et contrôle , Athérosclérose/microbiologie , Telomerase/métabolisme , Fragments peptidiques/pharmacologie , Maladie d'Alzheimer/métabolisme , Maladie d'Alzheimer/prévention et contrôle , Maladie d'Alzheimer/microbiologie , Parodontite/microbiologie , Parodontite/prévention et contrôle , Infections à Bacteroidaceae/microbiologie , Infections à Bacteroidaceae/complications , Infections à Bacteroidaceae/prévention et contrôle , Modèles animaux de maladie humaine , Souris de lignée C57BL , Mâle , Humains
12.
Biomolecules ; 14(6)2024 Jun 08.
Article de Anglais | MEDLINE | ID: mdl-38927075

RÉSUMÉ

Atherosclerosis (AS) has become the leading cause of cardiovascular disease worldwide. Our previous study had observed that Nippostrongylus brasiliensis (Nb) infection or its derived products could inhibit AS development by inducing an anti-inflammatory response. We performed a metabolic analysis to screen Nb-derived metabolites with anti-inflammation activity and evaluated the AS-prevention effect. We observed that the metabolite uridine had higher expression levels in mice infected with the Nb and ES (excretory-secretory) products and could be selected as a key metabolite. ES and uridine interventions could reduce the pro-inflammatory responses and increase the anti-inflammatory responses in vitro and in vivo. The apolipoprotein E gene knockout (ApoE-/-) mice were fed with a high-fat diet for the AS modeling. Following the in vivo intervention, ES products or uridine significantly reduced serum and liver lipid levels, alleviated the formation of atherosclerosis, and reduced the pro-inflammatory responses in serum or plaques, while the anti-inflammatory responses showed opposite trends. After blocking with 5-HD (5-hydroxydecanoate sodium) in vitro, the mRNA levels of M2 markers were significantly reduced. When blocked with 5-HD in vivo, the degree of atherosclerosis was worsened, the pro-inflammatory responses were increased compared to the uridine group, while the anti-inflammatory responses decreased accordingly. Uridine, a key metabolite from Nippostrongylus brasiliensis, showed anti-inflammatory and anti-atherosclerotic effects in vitro and in vivo, which depend on the activation of the mitochondrial ATP-sensitive potassium channel.


Sujet(s)
Anti-inflammatoires , Athérosclérose , Nippostrongylus , Uridine , Animaux , Mâle , Souris , Anti-inflammatoires/pharmacologie , Apolipoprotéines E/génétique , Apolipoprotéines E/déficit , Athérosclérose/métabolisme , Athérosclérose/génétique , Modèles animaux de maladie humaine , Canaux KATP/métabolisme , Canaux KATP/génétique , Souris knockout , Mitochondries/métabolisme , Mitochondries/effets des médicaments et des substances chimiques , Uridine/pharmacologie
13.
FASEB J ; 38(13): e23765, 2024 Jul 15.
Article de Anglais | MEDLINE | ID: mdl-38934372

RÉSUMÉ

The importance of autophagy in atherosclerosis has garnered significant attention regarding the potential applications of autophagy inducers. However, the impact of TAT-Beclin1, a peptide inducer of autophagy, on the development of atherosclerotic plaques remains unclear. Single-cell omics analysis indicates a notable reduction in GAPR1 levels within fibroblasts, stromal cells, and macrophages during atherosclerosis. Tat-beclin1 (T-B), an autophagy-inducing peptide derived from Beclin1, could selectively bind to GAPR1, relieving its inhibition on Beclin1 and thereby augmenting autophagosome formation. To investigate its impact on atherosclerosic plaque progression, we established the ApoE-/- mouse model of carotid atherosclerotic plaques. Surprisingly, intravenous administration of Tat-beclin1 dramatically accelerated the development of carotid artery plaques. Immunofluorescence analysis suggested that macrophage aggregation and autophagosome formation within atherosclerotic plaques were significantly increased upon T-B treatment. However, immunofluorescence and transmission electron microscopy (TEM) analysis revealed a reduction in autophagy flux through lysosomes. In vitro, the interaction between T-B and GAPR1 was confirmed in RAW264.7 cells, resulting in the increased accumulation of p62/SQSTM1 and LC3-II in the presence of ox-LDL. Additionally, T-B treatment elevated the protein levels of p62/SQSTM1, LC3-II, and cleaved caspase 1, along with the secretion of IL-1ß in response to ox-LDL exposure. In summary, our study underscores that T-B treatment amplifies abnormal autophagy and inflammation, consequently exacerbating atherosclerotic plaque development in ApoE-/- mice.


Sujet(s)
Apolipoprotéines E , Athérosclérose , Autophagie , Bécline-1 , Plaque d'athérosclérose , Animaux , Souris , Bécline-1/métabolisme , Bécline-1/génétique , Apolipoprotéines E/métabolisme , Apolipoprotéines E/déficit , Athérosclérose/métabolisme , Athérosclérose/anatomopathologie , Autophagie/effets des médicaments et des substances chimiques , Plaque d'athérosclérose/métabolisme , Plaque d'athérosclérose/anatomopathologie , Cellules RAW 264.7 , Souris de lignée C57BL , Mâle , Souris knockout , Macrophages/métabolisme
14.
Cell Signal ; 121: 111252, 2024 Sep.
Article de Anglais | MEDLINE | ID: mdl-38852936

RÉSUMÉ

BACKGROUND AND AIMS: S1P is an important factor regulating the function of the vascular endothelial barrier. SphK1 is an important limiting enzyme for the synthesis of S1P. However, the role of the SphK1/S1P-mediated vascular endothelial barrier function in atherosclerosis has not been fully revealed. This study explored the roles and mechanisms of SphK1 on atherosclerosis in vivo and in vitro. METHODS: In vivo, ApoE-/- and SphK1-/-ApoE-/- mice were fed a high-fat diet to induce atherosclerosis. In vitro, ox-LDL induced HUVECs to establish a cell model. Aortic histological changes were measured by H&E staining, Oil Red O staining, EVG staining, Sirius scarlet staining, immunofluorescence, and Evans Blue Assay. Western blotting was performed to explore the specific mechanism. RESULTS: We validated that deficiency of SphK1 resulted in a marked amelioration of atherosclerosis, as indicated by the decreased lipid accumulation, inflammatory factors, oxidative stress, aortic plaque area, inflammatory factor infiltration, VCAM-1 expression, and vascular endothelial permeability. Moreover, deficiency of SphK1 downregulated the expression of aortic S1PR3, Rhoa, ROCK, and F-actin. The results of administration with the SphK1 inhibitor PF-543 and the S1PR3 inhibitor VPC23019 in vitro further confirmed the conclusion that deficiency of SphK1 reduced S1P level and S1PR3 protein expression, inhibited Rhoa/ROCK signaling pathway, regulated protein expression of F-actin, improved vascular endothelial dysfunction and permeability, and exerted anti-atherosclerotic effects. CONCLUSIONS: This study revealed that deficiency of SphK1 relieved vascular endothelial barrier function in atherosclerosis mice via SphK1/S1P/S1PR signaling pathway.


Sujet(s)
Athérosclérose , Cellules endothéliales de la veine ombilicale humaine , Phosphotransferases (Alcohol Group Acceptor) , Transduction du signal , Récepteurs de la sphingosine-1-phosphate , rho-Associated Kinases , Protéine G RhoA , Animaux , Humains , Mâle , Souris , Apolipoprotéines E/déficit , Apolipoprotéines E/métabolisme , Apolipoprotéines E/génétique , Athérosclérose/métabolisme , Athérosclérose/anatomopathologie , Alimentation riche en graisse , Cellules endothéliales de la veine ombilicale humaine/métabolisme , Lysophospholipides/métabolisme , Souris de lignée C57BL , Souris knockout , Phosphotransferases (Alcohol Group Acceptor)/métabolisme , Phosphotransferases (Alcohol Group Acceptor)/génétique , Récepteurs aux lysosphingolipides/métabolisme , rho-Associated Kinases/métabolisme , Protéine G RhoA/métabolisme , Sphingosine/analogues et dérivés , Sphingosine/métabolisme , Récepteurs de la sphingosine-1-phosphate/métabolisme
15.
Sci Rep ; 14(1): 12450, 2024 05 30.
Article de Anglais | MEDLINE | ID: mdl-38816571

RÉSUMÉ

The effects of low doses of ionizing radiation on atherosclerosis remain uncertain, particularly as regards the generation of pro- or anti-inflammatory responses, and the time scale at which such effects can occur following irradiation. To explore these phenomena, we exposed atheroprone ApoE(-/-) mice to a single dose of 0, 0.05, 0.5 or 1 Gy of 137Cs (γ) administered at a 10.35 mGy min-1 dose rate and evaluated short-term (1-10 days) and long-term consequences (100 days). Bone marrow-derived macrophages were derived from mice 1 day after exposure. Irradiation was associated with a significant skewing of M0 and M2 polarized macrophages towards the M2 phenotype, as demonstrated by an increased mRNA expression of Retnla, Arg1, and Chil3 in cells from mice exposed to 0.5 or 1 Gy compared with non-irradiated animals. Minimal effects were noted in M1 cells or M1 marker mRNA. Concurrently, we observed a reduced secretion of IL-1ß but enhanced IL-10 release from M0 and M2 macrophages. Effects of irradiation on circulating monocytes were most marked at day 10 post-exposure, when the 1 Gy dose was associated with enhanced numbers of both Ly6CHigh and Ly6Low cells. By day 100, levels of circulating monocytes in irradiated and non-irradiated mice were equivalent, but anti-inflammatory Ly6CLow monocytes were significantly increased in the spleen of mice exposed to 0.05 or 1 Gy. Long term exposures did not affect atherosclerotic plaque size or lipid content, as determined by Oil red O staining, whatever the dose applied. Similarly, irradiation did not affect atherosclerotic plaque collagen or smooth muscle cell content. However, we found that lesion CD68+ cell content tended to decrease with rising doses of radioactivity exposure, culminating in a significant reduction of plaque macrophage content at 1 Gy. Taken together, our results show that short- and long-term exposures to low to moderate doses of ionizing radiation drive an anti-inflammatory response, skewing bone marrow-derived macrophages towards an IL-10-secreting M2 phenotype and decreasing plaque macrophage content. These results suggest a low-grade athero-protective effect of low and moderate doses of ionizing radiation.


Sujet(s)
Apolipoprotéines E , Radio-isotopes du césium , Rayons gamma , Macrophages , Plaque d'athérosclérose , Animaux , Macrophages/métabolisme , Macrophages/effets des radiations , Plaque d'athérosclérose/anatomopathologie , Plaque d'athérosclérose/métabolisme , Souris , Apolipoprotéines E/génétique , Apolipoprotéines E/déficit , Antigènes de différenciation des myélomonocytes/métabolisme , Antigènes CD/métabolisme , Antigènes CD/génétique , Athérosclérose/métabolisme , Athérosclérose/anatomopathologie , Mâle , Souris knockout ,
16.
Biochem Pharmacol ; 225: 116314, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38797271

RÉSUMÉ

Atherosclerosis, a chronic inflammatory disease, is the most relevant cause of carotid artery stenosis. Vascular endothelial cells (ECs) play a significant role in the development of atherosclerosis. In this chronic inflammatory environment, we aimed to investigate whether PCSK9 could mitigate atherosclerosis progression by reducing tissue factor expression in ECs via in vivo and in vitro assays. In vivo, we investigated the effect of PCSK9 inhibition on preventing atherosclerotic lesion formation in ApoE-/- mice fed a western diet. The results showed that inhibiting PCSK9 could significantly downregulate the protein expression of tissue factor (TF) in ECs to reduce the area of atherosclerotic plaques. In vitro, we incubated human umbilical vein endothelial cells (HUVECs) with lipopolysaccharide (LPS). We found that LPS-induced TF elevation was suppressed by a PCSK9 inhibitor at both the mRNA and protein levels and that the TLR4/NF-κB pathway was also suppressed by a PCSK9 inhibitor. With respect to plasma samples from patients with carotid artery stenosis, we also demonstrated that the expression of TF was positively correlated with that of PCSK9. Thus, in addition to regulating lipid metabolism, the regulation of endothelial cell TF expression through the TLR4/NF-κB pathway may be a potential mechanism of PCSK9 in promoting atherosclerotic carotid stenosis.


Sujet(s)
Apolipoprotéines E , Sténose carotidienne , Cellules endothéliales de la veine ombilicale humaine , Souris de lignée C57BL , Facteur de transcription NF-kappa B , Proprotéine convertase 9 , Transduction du signal , Thromboplastine , Récepteur de type Toll-4 , Animaux , Récepteur de type Toll-4/métabolisme , Récepteur de type Toll-4/génétique , Souris , Facteur de transcription NF-kappa B/métabolisme , Proprotéine convertase 9/génétique , Proprotéine convertase 9/métabolisme , Humains , Sténose carotidienne/métabolisme , Mâle , Apolipoprotéines E/génétique , Apolipoprotéines E/métabolisme , Apolipoprotéines E/déficit , Cellules endothéliales de la veine ombilicale humaine/métabolisme , Thromboplastine/métabolisme , Thromboplastine/génétique , Thromboplastine/biosynthèse , Transduction du signal/physiologie , Souris invalidées pour les gènes ApoE , Cellules endothéliales/métabolisme , Cellules endothéliales/effets des médicaments et des substances chimiques , Souris knockout , Inhibiteurs de PCSK9 , Femelle
17.
J Mol Med (Berl) ; 102(7): 887-897, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38733386

RÉSUMÉ

Atherosclerosis (AS) is a chronic inflammatory arterial disease, in which abnormal lipid metabolism and foam cell formation play key roles. Histamine is a vital biogenic amine catalyzed by histidine decarboxylase (HDC) from L-histidine. Histamine H1 receptor (H1R) antagonist is a commonly encountered anti-allergic agent in the clinic. However, the role and mechanism of H1R in atherosclerosis have not been fully elucidated. Here, we explored the effect of H1R on atherosclerosis using Apolipoprotein E-knockout (ApoE-/-) mice with astemizole (AST, a long-acting H1R antagonist) treatment. The results showed that AST increased atherosclerotic plaque area and hepatic lipid accumulation in mice. The result of microarray study identified a significant change of endothelial lipase (LIPG) in CD11b+ myeloid cells derived from HDC-knockout (HDC-/-) mice compared to WT mice. Blocking H1R promoted the formation of foam cells from bone marrow-derived macrophages (BMDMs) of mice by up-regulating p38 mitogen-activated protein kinase (p38 MAPK) and LIPG signaling pathway. Taken together, these findings demonstrate that blocking H1R signal aggravates atherosclerosis by promoting abnormal lipid metabolism and macrophage-derived foam cell formation via p38 MAPK-LIPG signaling pathway. KEY MESSAGES: Blocking H1R signal with AST aggravated atherosclerosis and increased hepatic lipid accumulation in high-fat diet (HFD)-fed ApoE-/- mice. Blocking H1R signal promoted macrophage-derived foam cell formation via p38 MAPK-LIPG signaling pathway.


Sujet(s)
Athérosclérose , Cellules spumeuses , Inflammation , Souris knockout , Récepteur histaminergique H1 , Transduction du signal , Animaux , Cellules spumeuses/métabolisme , Cellules spumeuses/anatomopathologie , Athérosclérose/métabolisme , Athérosclérose/étiologie , Athérosclérose/anatomopathologie , Souris , Inflammation/métabolisme , Inflammation/anatomopathologie , Récepteur histaminergique H1/métabolisme , Récepteur histaminergique H1/génétique , Mâle , Apolipoprotéines E/déficit , Apolipoprotéines E/génétique , Apolipoprotéines E/métabolisme , Souris de lignée C57BL , Métabolisme lipidique , p38 Mitogen-Activated Protein Kinases/métabolisme , Modèles animaux de maladie humaine , Plaque d'athérosclérose/métabolisme , Plaque d'athérosclérose/anatomopathologie , Alimentation riche en graisse/effets indésirables
18.
Article de Anglais | MEDLINE | ID: mdl-38780292

RÉSUMÉ

ABSTRACT: Arteriosclerosis (AS) is a chronic inflammatory disease and Buyang Huanwu decoction (BHD) has been identified as an anti-atherosclerosis effect, and the study is aimed to investigate the underlying mechanism. The E4 allele of Apolipoprotein E (ApoE) is associated with both metabolic dysfunction and an enhanced pro-inflammatory response, ApoE-knockout (ApoE-/-) mice were fed with a high-fat diet to establish an arteriosclerosis model and treated with BHD or atorvastatin (as a positive control). The atherosclerotic plaque in each mouse was evaluated using Oil red O Staining. Elisa kits were used to evaluate blood lipid, interleukin-6 (IL-6), IL-1 beta (IL-1ß), tumor necrosis factor alpha (TNF-α), IL-4, IL-10, and tumor growth factor beta (TGF-ß) contents, while Western blot was applicated to measure inducible nitric oxide synthase (iNOS), arginase I (Arg-1) expression. Meanwhile, pyruvate kinase M2 (PKM2), hypoxia-inducible factor-1 alpha (HIF-1α) and its target genes glucose transporter type 1 (GLUT1), lactate dehydrogenase A (LDHA), and 3-phosphoinositide-dependent kinase 1 (PDK1), as well as IL-6, IL-1ß, TNF-α, IL-4, IL-10, and TGF-ß were evaluated by the quantitative reverse transcription-polymerase chain reaction. BHD treatment significantly reduced body weight and arteriosclerosis plaque area and blood lipid levels including total cholesterol (TC), triglyceride (TG), low-density lipoprotein cholesterol (LDL-C), and high-density lipoprotein cholesterol (HDL-C). Meanwhile, BHD demonstrated a significant suppression of M1 polarization, by decreased secretion of iNOS and pro-inflammatory factors (IL-6, IL-1ß, and TNF-α) in ApoE-/- mice. The present study also revealed that BHD promotes the activation of M2 polarization, characterized by the expression of Arg-1 and anti-inflammatory factors (IL-4 and IL-10). In addition, PKM2/HIF-1α signaling was improved by M1/M2 macrophages polarization induced by BHD. The downstream target genes (GLUT1, LDHA, and PDK1) expression was significantly increased in high fat feeding ApoE-/- mice, and those of which were recused by BHD and Atorvastatin. These results suggested that M1/M2 macrophages polarization produce the inflammatory response against AS progress after BHD exposure.


Sujet(s)
Athérosclérose , Médicaments issus de plantes chinoises , Macrophages , Animaux , Médicaments issus de plantes chinoises/pharmacologie , Médicaments issus de plantes chinoises/usage thérapeutique , Souris , Athérosclérose/traitement médicamenteux , Athérosclérose/anatomopathologie , Athérosclérose/métabolisme , Macrophages/effets des médicaments et des substances chimiques , Macrophages/métabolisme , Mâle , Apolipoprotéines E/génétique , Apolipoprotéines E/déficit , Modèles animaux de maladie humaine , Souris invalidées pour les gènes ApoE , Souris knockout , Souris de lignée C57BL , Cytokines/métabolisme
19.
Article de Anglais | MEDLINE | ID: mdl-38780293

RÉSUMÉ

ABSTRACT: The traditional Chinese herbal prescription Buyang Huanwu decoction (BHD), effectively treats atherosclerosis. However, the mechanism of BHD in atherosclerosis remains unclear. We aimed to determine whether BHD could alleviate atherosclerosis by altering the microbiome-associated metabolic changes in atherosclerotic mice. An atherosclerotic model was established in apolipoprotein E-deficient mice fed high-fat diet, and BHD was administered through gavage for 12 weeks at 8.4 g/kg/d and 16.8 g/kg/d. The atherosclerotic plaque size, composition, serum lipid profile, and inflammatory cytokines, were assessed. Mechanistically, metabolomic and microbiota profiles were analyzed by liquid chromatography-mass spectrometry and 16S rRNA gene sequencing, respectively. Furthermore, intestinal microbiota and atherosclerosis-related metabolic parameters were correlated using Spearman analysis. Atherosclerotic mice treated with BHD exhibited reduced plaque area, aortic lumen occlusion, and lipid accumulation in the aortic root. Nine perturbed serum metabolites were significantly restored along with the relative abundance of microbiota at the family and genus levels but not at the phylum level. Gut microbiome improvement was strongly negatively correlated with improved metabolite levels. BHD treatment effectively slows the progression of atherosclerosis by regulating altered intestinal microbiota and perturbed metabolites.


Sujet(s)
Apolipoprotéines E , Athérosclérose , Alimentation riche en graisse , Médicaments issus de plantes chinoises , Microbiome gastro-intestinal , Animaux , Microbiome gastro-intestinal/effets des médicaments et des substances chimiques , Médicaments issus de plantes chinoises/pharmacologie , Athérosclérose/traitement médicamenteux , Athérosclérose/anatomopathologie , Athérosclérose/métabolisme , Alimentation riche en graisse/effets indésirables , Souris , Mâle , Apolipoprotéines E/déficit , Apolipoprotéines E/génétique , Apolipoprotéines E/métabolisme , Souris de lignée C57BL , Plaque d'athérosclérose/traitement médicamenteux , Plaque d'athérosclérose/anatomopathologie , Souris knockout , Souris invalidées pour les gènes ApoE
20.
Atherosclerosis ; 392: 117519, 2024 May.
Article de Anglais | MEDLINE | ID: mdl-38581737

RÉSUMÉ

BACKGROUND AND AIMS: Atherosclerosis is the primary underlying cause of myocardial infarction and stroke, which are the major causes of death globally. Heparanase (Hpse) is a pro-inflammatory extracellular matrix degrading enzyme that has been implicated in atherogenesis. However, to date the precise roles of Hpse in atherosclerosis and its mechanisms of action are not well defined. This study aims to provide new insights into the contribution of Hpse in different stages of atherosclerosis in vivo. METHODS: We generated Hpse gene-deficient mice on the atherosclerosis-prone apolipoprotein E gene knockout (ApoE-/-) background to investigate the impact of Hpse gene deficiency on the initiation and progression of atherosclerosis after 6 and 14 weeks high-fat diet feeding, respectively. Atherosclerotic lesion development, blood serum profiles, lesion composition and aortic immune cell populations were evaluated. RESULTS: Hpse-deficient mice exhibited significantly reduced atherosclerotic lesion burden in the aortic sinus and aorta at both time-points, independent of changes in plasma cholesterol levels. A significant reduction in the necrotic core size and an increase in smooth muscle cell content were also observed in advanced atherosclerotic plaques of Hpse-deficient mice. Additionally, Hpse deficiency reduced circulating and aortic levels of VCAM-1 at the initiation and progression stages of disease and circulating MCP-1 levels in the initiation but not progression stage. Moreover, the aortic levels of total leukocytes and dendritic cells in Hpse-deficient ApoE-/- mice were significantly decreased compared to control ApoE-/-mice at both disease stages. CONCLUSIONS: This study identifies Hpse as a key pro-inflammatory enzyme driving the initiation and progression of atherosclerosis and highlighting the potential of Hpse inhibitors as novel anti-inflammatory treatments for cardiovascular disease.


Sujet(s)
Aorte , Athérosclérose , Glucuronidase , Souris invalidées pour les gènes ApoE , Plaque d'athérosclérose , Animaux , Mâle , Souris , Aorte/anatomopathologie , Aorte/métabolisme , Aorte/enzymologie , Maladies de l'aorte/anatomopathologie , Maladies de l'aorte/génétique , Maladies de l'aorte/enzymologie , Maladies de l'aorte/métabolisme , Apolipoprotéines E/génétique , Apolipoprotéines E/déficit , Athérosclérose/génétique , Athérosclérose/anatomopathologie , Athérosclérose/enzymologie , Athérosclérose/métabolisme , Alimentation riche en graisse , Modèles animaux de maladie humaine , Évolution de la maladie , Glucuronidase/déficit , Glucuronidase/génétique , Glucuronidase/métabolisme , Souris de lignée C57BL , Souris knockout , Nécrose , Sinus de l'aorte/anatomopathologie , Molécule-1 d'adhérence des cellules vasculaires/métabolisme
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE