Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 11.185
Filtrer
1.
PeerJ ; 12: e17915, 2024.
Article de Anglais | MEDLINE | ID: mdl-39184397

RÉSUMÉ

Oxidized light-density lipoprotein (ox-LDL) causes endothelial dysfunction, which is an important determinant of atherogenesis, and subsequently leads to apoptosis. Atherosclerosis is one of the most significant cardiovascular diseases (CVDs) threatening human health and causes death worldwide. Recently, long noncoding RNAs (lncRNAs) have been suggested to involved in vascular biology. Ox-LDL activates nuclear factor kappa-B (NF-κB), and NF-κB interacting lncRNA (NKILA) inhibits NF-κB signaling. In this study, the hypothesis is that NKILA may regulate endothelial cell (EC) apoptosis and, therefore, play a role in the pathogenesis of atherosclerosis. This hypothesis is based on the knowledge that EC apoptosis contributes to atherosclerosis development and that NKILA has become a prominent lncRNA in CVDs. The expression of Bcl-2-associated X protein (BAX), caspase 9 (CASP9), cytochrome c (Cyt c, CYCS), apoptotic protease activating factor 1 (APAF1), and B-cell lymphoma 2 (BCL-2) genes in human umbilical vein endothelial cells (HUVEC) treated with ox-LDL and transfected with NKILA siRNA was analyzed using quantitative reverse transcription polymerase chain reaction (RT-qPCR). BAX, CASP9, CYCS, APAF1, and BCL-2 gene expression was downregulated in ox-LDL and NKILA siRNA-treated HUVEC. In addition, when threshold/quantification cycle (Cq) values of NKILA gene expression increased, Cq values of BAX, CASP9, APAF1, and BCL-2 gene expression increased statistics significantly. The expression detection of all these genes, resulting from NKILA gene silencing, may provide guidance for epigenetic studies on EC apoptosis in atherosclerosis.


Sujet(s)
Apoptose , Facteur-1 activateur des protéases apoptotiques , Athérosclérose , Cellules endothéliales de la veine ombilicale humaine , ARN long non codant , Humains , Athérosclérose/génétique , Athérosclérose/métabolisme , Athérosclérose/anatomopathologie , Apoptose/génétique , ARN long non codant/génétique , ARN long non codant/métabolisme , Facteur-1 activateur des protéases apoptotiques/génétique , Facteur-1 activateur des protéases apoptotiques/métabolisme , Lipoprotéines LDL/métabolisme , Caspase-9/génétique , Caspase-9/métabolisme , Protéine Bax/génétique , Protéine Bax/métabolisme , Cytochromes c/métabolisme , Cytochromes c/génétique , Protéines proto-oncogènes c-bcl-2/génétique , Protéines proto-oncogènes c-bcl-2/métabolisme , Régulation de l'expression des gènes
2.
Bull Math Biol ; 86(9): 112, 2024 Aug 02.
Article de Anglais | MEDLINE | ID: mdl-39093509

RÉSUMÉ

Macrophages in atherosclerotic lesions exhibit a spectrum of behaviours or phenotypes. The phenotypic distribution of monocyte-derived macrophages (MDMs), its correlation with MDM lipid content, and relation to blood lipoprotein densities are not well understood. Of particular interest is the balance between low density lipoproteins (LDL) and high density lipoproteins (HDL), which carry bad and good cholesterol respectively. To address these issues, we have developed a mathematical model for early atherosclerosis in which the MDM population is structured by phenotype and lipid content. The model admits a simpler, closed subsystem whose analysis shows how lesion composition becomes more pathological as the blood density of LDL increases relative to the HDL capacity. We use asymptotic analysis to derive a power-law relationship between MDM phenotype and lipid content at steady-state. This relationship enables us to understand why, for example, lipid-laden MDMs have a more inflammatory phenotype than lipid-poor MDMs when blood LDL lipid density greatly exceeds HDL capacity. We show further that the MDM phenotype distribution always attains a local maximum, while the lipid content distribution may be unimodal, adopt a quasi-uniform profile or decrease monotonically. Pathological lesions exhibit a local maximum in both the phenotype and lipid content MDM distributions, with the maximum at an inflammatory phenotype and near the lipid content capacity respectively. These results illustrate how macrophage heterogeneity arises in early atherosclerosis and provide a framework for future model validation through comparison with single-cell RNA sequencing data.


Sujet(s)
Athérosclérose , Lipoprotéines HDL , Lipoprotéines LDL , Macrophages , Concepts mathématiques , Phénotype , Humains , Macrophages/métabolisme , Macrophages/anatomopathologie , Athérosclérose/anatomopathologie , Athérosclérose/métabolisme , Athérosclérose/sang , Lipoprotéines LDL/métabolisme , Lipoprotéines LDL/sang , Lipoprotéines HDL/sang , Lipoprotéines HDL/métabolisme , Modèles cardiovasculaires , Métabolisme lipidique , Lipoprotéines/métabolisme , Lipoprotéines/sang , Simulation numérique
3.
FASEB J ; 38(15): e23857, 2024 Aug 15.
Article de Anglais | MEDLINE | ID: mdl-39114953

RÉSUMÉ

Atherosclerotic plaque formation is largely attributed to the impaired efferocytosis, which is known to be associated with the pathologic upregulation of cluster of differentiation 47 (CD47), a key antiphagocytic molecule. By gene expression omnibus (GEO) datasets analysis, we identified that four miRNAs are aberrantly downregulated in atherosclerosis, coronary artery disease, and obesity. Of them, hsa-miR-299-3p (miR-299-3p) was predicted to target the 3'UTR of human CD47 mRNA by bioinformatics analysis. Further, we demonstrated that miR-299-3p negatively regulates CD47 expression by binding to the target sequence "CCCACAU" in the 3'UTR of CD47 mRNA through luciferase reporter assay and site-directed mutagenesis. Additionally, we found that miR-299-3p was downregulated by ~32% in foam cells in response to oxidized low-density lipoprotein (ox-LDL) stimulation, thus upregulating CD47 and contributing to the impaired efferocytosis. Whereas, restoration of miR-299-3p reversed the ox-LDL-induced upregulation of CD47, thereby facilitating efferocytosis. In high-fat diet (HFD) fed ApoE-/- mice, we discovered that miR-299-3p was downregulated thus leading to upregulation of CD47 in abdominal aorta. Conversely, miR-299-3p restoration potently suppressed HFD-induced upregulation of CD47 and promoted phagocytosis of foam cells by macrophages in atherosclerotic plaques, thereby reducing necrotic core, increasing plaque stability, and mitigating atherosclerosis. Conclusively, we identify miR-299-3p as a negative regulator of CD47, and reveal a molecular mechanism whereby the ox-LDL-induced downregulation of miR-299-3p leads to the upregulation of CD47 in foam cells thus contributing to the impaired efferocytosis in atherosclerosis, and propose miR-299-3p can potentially serve as an inhibitor of CD47 to promote efferocytosis and ameliorate atherosclerosis.


Sujet(s)
Athérosclérose , Antigènes CD47 , , microARN , Animaux , Humains , Souris , Régions 3' non traduites , Athérosclérose/métabolisme , Athérosclérose/génétique , Athérosclérose/anatomopathologie , Antigènes CD47/métabolisme , Antigènes CD47/génétique , Alimentation riche en graisse/effets indésirables , Cellules spumeuses/métabolisme , Cellules spumeuses/anatomopathologie , Lipoprotéines LDL/métabolisme , Souris de lignée C57BL , microARN/génétique , microARN/métabolisme
4.
Mol Med ; 30(1): 117, 2024 Aug 09.
Article de Anglais | MEDLINE | ID: mdl-39123116

RÉSUMÉ

BACKGROUND: Lipid metabolism disorders are associated with degeneration of multiple tissues and organs, but the mechanism of crosstalk between lipid metabolism disorder and intervertebral disc degeneration (IDD) has not been fully elucidated. In this study we aim to investigate the regulatory mechanism of abnormal signal of lipid metabolism disorder on intervertebral disc endplate chondrocyte (EPC) senescence and calcification. METHODS: Human intervertebral disc cartilage endplate tissue, cell model and rat hyperlipemia model were performed in this study. Histology and immunohistochemistry were used to human EPC tissue detection. TMT-labelled quantitative proteomics was used to detect differential proteins, and MRI, micro-CT, safranin green staining and immunofluorescence were performed to observe the morphology and degeneration of rat tail intervertebral discs. Flow cytometry, senescence-associated ß-galactosidase staining, alizarin red staining, alkaline phosphatase staining, DCFH-DA fluorescent probe, and western blot were performed to detect the expression of EPC cell senescence, senescence-associated secretory phenotype, calcification-related proteins and the activation of cell senescence-related signaling pathways. RESULTS: Our study found that the highly expressed oxidized low-density lipoprotein (ox-LDL) and Lectin-like oxidized low-density lipoprotein receptor 1 (LOX-1) in human degenerative EPC was associated with hyperlipidemia (HLP). TMT-labelled quantitative proteomics revealed enriched pathways such as cell cycle regulation, endochondral bone morphogenesis and inflammation. The rat model revealed that HLP could induce ox-LDL, LOX-1, senescence and calcification markers high expression in EPC. Moreover, we demonstrated that ox-LDL-induced EPCs senescence and calcification were dependent on the LOX-1 receptor, and the ROS/P38-MAPK/NF-κB signaling pathway was implicated in the regulation of senescence induced by ox-LDL/LOX-1 in cell model. CONCLUSIONS: So our study revealed that ox-LDL/LOX-1-induced EPCs senescence and calcification through ROS/P38-MAPK/NF-κB signaling pathway, providing information on understanding the link between lipid metabolism disorders and IDD.


Sujet(s)
Vieillissement de la cellule , Chondrocytes , Dégénérescence de disque intervertébral , Métabolisme lipidique , Lipoprotéines LDL , Récepteurs éboueurs de classe E , Dégénérescence de disque intervertébral/métabolisme , Dégénérescence de disque intervertébral/anatomopathologie , Lipoprotéines LDL/métabolisme , Animaux , Humains , Récepteurs éboueurs de classe E/métabolisme , Chondrocytes/métabolisme , Chondrocytes/anatomopathologie , Rats , Mâle , Calcinose/métabolisme , Calcinose/anatomopathologie , Disque intervertébral/métabolisme , Disque intervertébral/anatomopathologie , Modèles animaux de maladie humaine , Femelle , Adulte d'âge moyen , Transduction du signal , Adulte , Protéomique/méthodes , Rat Sprague-Dawley
5.
Discov Med ; 36(187): 1721-1731, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-39190387

RÉSUMÉ

BACKGROUND: Hyperlipidemia is one of the main causes of aggravated hepatic ischemia-reperfusion injury (IRI). Simvastatin (SIM), a lipid-lowering drug, has been shown to effectively alleviate IRI caused by hyperlipidemia. However, the regulatory mechanism by which SIM alleviates hyperlipidemia-induced hepatic IRI is still not clear. This study aims to explore the potential mechanisms of SIM in inhibiting hyperlipidemia-induced hepatic IRI, providing new therapeutic strategies for the alleviation of hepatic IRI. METHODS: An animal model of hyperlipidemia was induced by feeding mice a high-fat diet for 8 weeks. Subsequently, a hepatic IRI animal model of hyperlipidemia was established by occluding the hepatic artery and portal vein for one hour, followed by reperfusion for 6 or 12 h. Enzyme linked immunosorbent assay, Western blotting, hematoxylin-eosin (H&E) staining, immunohistochemistry, immunofluorescence, and Terminal-deoxynucleoitidyl Transferase Mediated Nick End Labeling assay, were used to evaluate liver injury, neutrophil extracellular traps (NETs) formation, and related molecular mechanisms. RESULTS: Hepatic IRI was accelerated by hyperlipidemia, which enhanced the expression of oxidized low-density lipoprotein (oxLDL) and Macrophage-1antigen (Mac-1), leading to the promotion of NETs formation and apoptosis of liver cells. The administration of simvastatin reduced the levels of oxLDL and Mac-1, decreased the formation of NETs, and alleviated hepatic IRI induced by hyperlipidemia. CONCLUSIONS: Simvastatin reduced hyperlipidemia-induced hepatic IRI by inhibiting the formation of NETs through the regulation of the oxLDL/Mac-1 pathway.


Sujet(s)
Alimentation riche en graisse , Pièges extracellulaires , Hyperlipidémies , Foie , Lésion d'ischémie-reperfusion , Simvastatine , Animaux , Simvastatine/pharmacologie , Simvastatine/usage thérapeutique , Lésion d'ischémie-reperfusion/prévention et contrôle , Lésion d'ischémie-reperfusion/anatomopathologie , Lésion d'ischémie-reperfusion/traitement médicamenteux , Lésion d'ischémie-reperfusion/métabolisme , Souris , Pièges extracellulaires/effets des médicaments et des substances chimiques , Pièges extracellulaires/métabolisme , Mâle , Alimentation riche en graisse/effets indésirables , Foie/anatomopathologie , Foie/effets des médicaments et des substances chimiques , Foie/métabolisme , Hyperlipidémies/traitement médicamenteux , Hyperlipidémies/métabolisme , Hyperlipidémies/anatomopathologie , Hyperlipidémies/complications , Souris de lignée C57BL , Modèles animaux de maladie humaine , Lipoprotéines LDL/métabolisme , Transduction du signal/effets des médicaments et des substances chimiques , Apoptose/effets des médicaments et des substances chimiques
6.
Discov Med ; 36(187): 1678-1691, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-39190383

RÉSUMÉ

BACKGROUND: Remodeling of vascular smooth muscle cells (VSMCs), as a pathological hallmark of cardiovascular diseases, is related to the molecular rewiring of Calcium signaling, which induces upregulation of stromal interaction molecule (STIM) proteins. This study analyzed the influence of STIM1 proteins on the remodeling of VSMCs in atherosclerosis (AS). METHODS: After oxidized low-density lipoprotein (ox-LDL) treatment and transfection, VSMC viability, migration, and invasion were separately measured using Cell Counting Kit-8, Scratch assay, and Transwell assay. An animal AS model was constructed, and histological analysis via hematoxylin-eosin staining was conducted on the aorta. RESULTS: Ox-LDL promoted expression of STIM1 and Orai calcium release-activated calcium modulator 1 (Orai1). STIM1 or Orai1 downregulation suppressed viability, migration, invasion, and phenotypic switching of ox-LDL-treated VSMCs, whereas STIM1 or Orai1 upregulation had opposite effects. Orai1 level was upregulated by STIM1 overexpression. Orai1 silencing reversed the effects of STIM1 overexpression in VSMCs. STIM1 deficiency alleviated AS and regulated expression of Orai1 and phenotypic switch-related factors in vivo. CONCLUSION: STIM1 deficiency suppresses viability, migration, invasion, and phenotypic switching of ox-LDL-induced VSMCs and alleviates AS by inhibiting Orai1.


Sujet(s)
Athérosclérose , Mouvement cellulaire , Lipoprotéines LDL , Muscles lisses vasculaires , Myocytes du muscle lisse , Protéine ORAI1 , Molécule-1 d'interaction stromale , Molécule-1 d'interaction stromale/métabolisme , Molécule-1 d'interaction stromale/génétique , Athérosclérose/anatomopathologie , Athérosclérose/métabolisme , Muscles lisses vasculaires/anatomopathologie , Muscles lisses vasculaires/métabolisme , Animaux , Protéine ORAI1/métabolisme , Protéine ORAI1/génétique , Protéine ORAI1/antagonistes et inhibiteurs , Myocytes du muscle lisse/métabolisme , Myocytes du muscle lisse/anatomopathologie , Lipoprotéines LDL/métabolisme , Mouvement cellulaire/effets des médicaments et des substances chimiques , Humains , Mâle , Souris , Survie cellulaire/effets des médicaments et des substances chimiques , Modèles animaux de maladie humaine , Remodelage vasculaire/effets des médicaments et des substances chimiques , Protéines tumorales/métabolisme , Protéines tumorales/génétique
7.
Pediatr Rheumatol Online J ; 22(1): 73, 2024 Aug 08.
Article de Anglais | MEDLINE | ID: mdl-39118148

RÉSUMÉ

BACKGROUND: Juvenile dermatomyositis (JDM) is a systemic vasculopathy associated with metabolic derangements and possible increased risk for premature atherosclerosis. Oxidation of low-density lipoprotein (LDL) in the endothelium is an early step in atherosclerotic plaque formation. It is not known if oxidized LDL is altered in children with untreated JDM. The deposition of oxidized LDL in the vasculature of muscle biopsies (MBx) from patients with untreated JDM and pediatric controls was assessed. FINDINGS: Frozen tissue sections of MRI-directed MBx from 20 female children with untreated JDM and 5 female controls were stained with DAPI and fluorescently labeled antibodies against von Willebrand factor (vWF) and LDL oxidized by copper (oxLDL). Blood vessels were identified by positive vWF staining, and total fluorescence of oxLDL within the vessel walls was measured. Children with untreated JDM had increased deposition of oxLDL in the walls of muscle vasculature compared to healthy children (difference in means ± SEM = 19.86 ± 8.195, p = 0.03). Within the JDM cohort, there was a trend towards increased oxLDL deposition with longer duration of untreated disease (r = 0.43, p = 0.06). There was no significant correlation found between oxLDL deposition and markers of acute JDM disease activity including disease activity scores or muscle enzymes. CONCLUSIONS: This study found increased deposition of oxLDL within blood vessels of children with untreated JDM supporting the concern that these children are at increased risk for premature atherosclerosis from chronic exposure to vascular oxLDL. This study highlights the importance of early diagnosis and treatment initiation to ameliorate cardiovascular damage.


Sujet(s)
Dermatomyosite , Lipoprotéines LDL , Humains , Femelle , Lipoprotéines LDL/métabolisme , Dermatomyosite/métabolisme , Dermatomyosite/anatomopathologie , Enfant , Adolescent , Muscles squelettiques/métabolisme , Muscles squelettiques/vascularisation , Muscles squelettiques/anatomopathologie , Enfant d'âge préscolaire , Études cas-témoins , Imagerie par résonance magnétique/méthodes , Biopsie
8.
Immunity ; 57(7): 1448-1451, 2024 Jul 09.
Article de Anglais | MEDLINE | ID: mdl-38986438

RÉSUMÉ

Autoreactive lymphocytes that infiltrate the pancreatic islet environment and target ß cells are primary drivers of type 1 diabetes. In this issue of Immunity, Srivastava et al.1 examine the role of the islet microenvironment in autoimmunity and find that the scavenging receptor CXCL16 on islet-resident macrophages uptakes oxidized low-density lipoproteins and promotes the differentiation and survival of infiltrating pathogenic CD8+ T cells.


Sujet(s)
Auto-immunité , Lymphocytes T CD8+ , Diabète de type 1 , Ilots pancréatiques , Macrophages , Auto-immunité/immunologie , Diabète de type 1/immunologie , Ilots pancréatiques/immunologie , Ilots pancréatiques/métabolisme , Humains , Animaux , Macrophages/immunologie , Macrophages/métabolisme , Lymphocytes T CD8+/immunologie , Cellules à insuline/immunologie , Cellules à insuline/métabolisme , Lipoprotéines LDL/métabolisme , Lipoprotéines LDL/immunologie
9.
Acta Biochim Pol ; 71: 13004, 2024.
Article de Anglais | MEDLINE | ID: mdl-39041003

RÉSUMÉ

CD36 is a type 2 cell surface scavenger receptor expressed in various tissues. In macrophages, CD36 recognizes oxidized low-density lipoprotein (ox-LDL), which promotes the formation of foam cells, the first step toward an atherosclerotic arterial lesion. CD36 possesses a variety of posttranslational modifications, among them N-glycosylation and O-GlcNAc modification. Some of the roles of these modifications on CD36 are known, such as N-linked glycosylation, which provides proper folding and trafficking to the plasma membrane in the human embryonic kidney. This study aimed to determine whether variations in the availability of UDP-GlcNAc could impact Rab-5-mediated endocytic trafficking and, therefore, the cellular localization of CD36. These preliminary results suggest that the availability of the substrate UDP-GlcNAc, modulated in response to treatment with Thiamet G (TMG), OSMI-1 (O-GlcNAcylation enzymes modulators) or Azaserine (HBP modulator), influences the localization of CD36 in J774 macrophages, and the endocytic trafficking as evidenced by the regulatory protein Rab-5, between the plasma membrane and the cytoplasm.


Sujet(s)
Antigènes CD36 , Macrophages , Antigènes CD36/métabolisme , Macrophages/métabolisme , Animaux , Souris , Lignée cellulaire , Glycosylation , Membrane cellulaire/métabolisme , Humains , Lipoprotéines LDL/métabolisme , Hexosamine/métabolisme , Hexosamine/biosynthèse , Protéines G rab5/métabolisme , Transport des protéines , Voies de biosynthèse , Maturation post-traductionnelle des protéines
10.
Int Heart J ; 65(4): 738-747, 2024.
Article de Anglais | MEDLINE | ID: mdl-39085113

RÉSUMÉ

The regulatory factor X7 (RFX7) is a vital mediator in atherosclerosis. This study aims to discuss the effect and underlying mechanism of RFX7 on the regulation of oxidized low-density lipoprotein (ox-LDL) -induced proliferation and migration of vascular smooth muscle cells (VSMCs).Ox-LDL was used to construct atherosclerosis in vitro model. The mRNA and protein levels of RFX7 and Sirtuin 4 (SIRT4) were evaluated by quantitative real-time polymerase chain reaction (qRT-PCR) or western blot assays. The cellular functions were measured via 3-[4,5-dimethylthiazol-2-yl]-2,5-diphenyl tetrazolium bromide (MTT), EdU, flow cytometry, and wound healing assay assays. The interaction between RFX7 and SIRT4 promoter was validated using chromatin immunoprecipitation and dual-luciferase reporter assays.The stimulation with ox-LDL elevated the viability of VSMCs and decreased the mRNA and protein levels of RFX7 and SIRT4 in VSMCs in a dose-dependent manner. Functionally, RFX7 overexpression restrained the VSMC viability, proliferation, and migration induced by ox-LDL, but facilitated VSMC apoptosis. RFX7 elevated SIRT4 expression via binding to its promoter. Furthermore, overexpressing either SIRT4 or RFX7 inactivated JAK2/STAT3 signaling, causing a decrease in VSMC proliferation and migration and an increase in VSMC apoptosis when exposed to ox-LDL. The impact of RFX7 overexpression on JAK2/STAT3 signaling and cellular function following ox-LDL exposure was abrogated by SIRT4 silencing.The heightened RFX7 expression restrained the proliferation and migration of ox-LDL-stimulated VSMCs via SIRT4-mediated inactivation of JAK2/STAT3 pathway.


Sujet(s)
Mouvement cellulaire , Prolifération cellulaire , Kinase Janus-2 , Lipoprotéines LDL , Muscles lisses vasculaires , Facteur de transcription STAT-3 , Transduction du signal , Sirtuines , Facteur de transcription STAT-3/métabolisme , Lipoprotéines LDL/pharmacologie , Lipoprotéines LDL/métabolisme , Kinase Janus-2/métabolisme , Muscles lisses vasculaires/métabolisme , Muscles lisses vasculaires/cytologie , Humains , Sirtuines/métabolisme , Sirtuines/génétique , Athérosclérose/métabolisme , Cellules cultivées , Myocytes du muscle lisse/métabolisme , Myocytes du muscle lisse/effets des médicaments et des substances chimiques , Protéines mitochondriales
11.
Cardiovasc Toxicol ; 24(8): 800-817, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-38951468

RÉSUMÉ

Radix Paeoniae Rubra. (Chishao, RPR) and Cortex Moutan. (Mudanpi, CM) are a pair of traditional Chinese medicines that play an important role in the treatment of atherosclerosis (AS). The main objective of this study was to identify potential synergetic function and underlying mechanisms of RPR-CM in the treatment of AS. The main active ingredients, targets of RPR-CM and AS-related genes were obtained from public databases. A Venn diagram was utilized to screen the common targets of RPR-CM in treating AS. The protein-protein interaction network was established based on STRING database. Biological functions and pathways of potential targets were analyzed through Gene Ontology and Kyoto Encyclopedia of Genes and Genomes enrichment analyses. Cytoscape was used to construct the drug-compound-target-signal pathway network. Molecular docking was performed to verify the binding ability of the bioactive ingredients and the target proteins. The endothelial inflammation model was constructed with human umbilical vein endothelial cells stimulated with ox-LDL, and the function of RPR-CM in treating AS was verified by CCK-8 assay, enzyme-linked immunosorbent assay, and qPCR. In this study, 12 active components and 401 potential target genes of RPR-CM were identified, among which quercetin, kaempferol and baicalein were considered to be the main active components. A total of 1903 AS-related genes were identified through public databases and four GEO datasets (GSE57691, GSE72633, GSE6088 and GSE199819). There are 113 common target genes of RPR-CM in treating AS. PPI network analysis identified 17 genes in cluster 1 as the core targets. Bioinformatics analysis showed that RPR-CM in AS treatment was associated with multiple downstream biological processes and signal pathways. PTGS2, JUN, CASP3, TNF, IL1B, IL6, FOS, STAT1 were identified as the core targets of RPR-CM, and molecular docking showed that the main bioactive components of RPR-CM had good binding ability with the core targets. RPR-CM extract significantly inhibited the levels of inflammatory factors TNF-α, IL-6, IL-1ß, MCP-1, VCAM-1 and ICAM-1 in HUVECs, and inhibited endothelial inflammation. This study revealed the active ingredients of RPR-CM, and identified the key downstream targets and signaling pathways in the treatment of AS, providing theoretical basis for the application of RPR-CM in prevention and treatment of AS.


Sujet(s)
Anti-inflammatoires , Athérosclérose , Médicaments issus de plantes chinoises , Cellules endothéliales de la veine ombilicale humaine , Simulation de docking moléculaire , Pharmacologie des réseaux , Paeonia , Cartes d'interactions protéiques , Transduction du signal , Humains , Cellules endothéliales de la veine ombilicale humaine/métabolisme , Cellules endothéliales de la veine ombilicale humaine/effets des médicaments et des substances chimiques , Médicaments issus de plantes chinoises/pharmacologie , Médicaments issus de plantes chinoises/composition chimique , Paeonia/composition chimique , Transduction du signal/effets des médicaments et des substances chimiques , Athérosclérose/traitement médicamenteux , Athérosclérose/métabolisme , Athérosclérose/génétique , Athérosclérose/anatomopathologie , Anti-inflammatoires/pharmacologie , Anti-inflammatoires/isolement et purification , Lipoprotéines LDL/métabolisme , Régulation de l'expression des gènes/effets des médicaments et des substances chimiques , Bases de données génétiques , Médiateurs de l'inflammation/métabolisme , Cellules cultivées , Cytokines/métabolisme , Cytokines/génétique , Réseaux de régulation génique
12.
Biomater Sci ; 12(16): 4181-4193, 2024 Aug 06.
Article de Anglais | MEDLINE | ID: mdl-38979569

RÉSUMÉ

Drug delivery systems based on biomimetic peptide nanoparticles are steadily gaining prominence in the treatment of diverse medical conditions. This study focused on the development of peptides that depend on ligand-receptor interactions to load rapamycin (RAPA). Furthermore, a multifunctional peptide was engineered to target oxidized low-density lipoprotein (oxLDL) within atherosclerotic plaques, facilitating the localized delivery of RAPA. The interactions between peptides and RAPA/oxLDL were analyzed by simulations and experimental approaches. Results show that the main amino acid residues on the mammalian target of rapamycin that bind to RAPA are constructed as peptides (P1 and P2), which have specific interactions with RAPA and can effectively improve the loading efficiency of RAPA. The encapsulation and drug loading efficiencies of P1/P2 were 68.0/47.9% and 48.3/36.5%, respectively. In addition, the interaction force of the multifunctional peptide (P3) and oxLDL surpassed that of their interaction with human umbilical vein endothelial cells by a factor of 3.6, conclusively establishing the specific targeting of oxLDL by these nanoparticles. The encapsulation and drug loading efficiencies of P3 for RAPA were determined to be 60.2% and 41.5%. P3 can effectively load RAPA and target oxLDL within the plaque, suggesting that P3 has potential as a therapeutic agent for atherosclerotic disease.


Sujet(s)
Cellules endothéliales de la veine ombilicale humaine , Lipoprotéines LDL , Nanoparticules , Peptides , Plaque d'athérosclérose , Sirolimus , Lipoprotéines LDL/composition chimique , Lipoprotéines LDL/métabolisme , Sirolimus/administration et posologie , Sirolimus/composition chimique , Sirolimus/pharmacologie , Humains , Plaque d'athérosclérose/traitement médicamenteux , Peptides/composition chimique , Peptides/pharmacologie , Peptides/administration et posologie , Nanoparticules/composition chimique , Nanoparticules/administration et posologie , Systèmes de délivrance de médicaments , Matériaux biomimétiques/composition chimique , Matériaux biomimétiques/pharmacologie , Vecteurs de médicaments/composition chimique
13.
Biosci Rep ; 44(8)2024 Aug 28.
Article de Anglais | MEDLINE | ID: mdl-39034849

RÉSUMÉ

OBJECTIVE: Estrogen receptor-positive (ER+) breast cancer represents about 80% of cases, tamoxifen is the election neoadjuvant chemotherapy. However, a large percentage of patients develop chemoresistance, compromising recovery. Clinical evidence suggests that high plasmatic levels of low-density lipoproteins (LDL) could promote cancer progression. The present study analyzed the effect of LDL on the primary plasmatic active Tamoxifen's metabolites resistance acquisition, 4-hydroxytamoxifen (4OH-Tam) and 4-hydroxy-N-desmethyl-tamoxifen (endoxifen), in breast cancer ERα + cells (MCF-7). METHODS: Two resistant cellular variants, MCF-7Var-H and MCF-7Var-I, were generated by a novel strategy and their phenotype features were evaluated. Phenotypic assessment was performed by MTT assays, cytometry, immunofluorescence microscopy, zymography and protein expression analysis. RESULTS: MCF-7Var-H, generated only with tamoxifen metabolites, showed a critical down-regulation in hormone receptors, augmented migration capacity, metalloprotease 9 extracellular medium excretion, and a mesenchymal morphology in contrast with native MCF-7, suggesting the transition towards Triple-negative breast cancer (TNBC) phenotype. In contrast, MCF-7Var-I which was generated in a high LDL media, showed only a slight upregulation in ER and other less noticeable metabolic adaptations. Results suggest a potential role of transcription factor nuclear factor erythroid 2-related factor 2 (Nrf2) in phenotypic differences observed among variants. CONCLUSION: LDL high or low concentrations during Tamoxifen´s metabolites chemoresistance acquisition leads to different cellular mechanisms related to chemoresistance. A novel adaptative cellular response associated with Nrf2 activity could be implicated.


Sujet(s)
Résistance aux médicaments antinéoplasiques , Récepteur alpha des oestrogènes , Lipoprotéines LDL , Phénotype , Tamoxifène , Tumeurs du sein triple-négatives , Humains , Tamoxifène/pharmacologie , Tamoxifène/analogues et dérivés , Résistance aux médicaments antinéoplasiques/effets des médicaments et des substances chimiques , Cellules MCF-7 , Femelle , Récepteur alpha des oestrogènes/métabolisme , Récepteur alpha des oestrogènes/génétique , Tumeurs du sein triple-négatives/traitement médicamenteux , Tumeurs du sein triple-négatives/métabolisme , Tumeurs du sein triple-négatives/anatomopathologie , Lipoprotéines LDL/métabolisme , Antinéoplasiques hormonaux/pharmacologie , Mouvement cellulaire/effets des médicaments et des substances chimiques , Tumeurs du sein/traitement médicamenteux , Tumeurs du sein/métabolisme , Tumeurs du sein/anatomopathologie , Régulation de l'expression des gènes tumoraux/effets des médicaments et des substances chimiques
14.
Aging (Albany NY) ; 16(13): 10784-10798, 2024 Jul 10.
Article de Anglais | MEDLINE | ID: mdl-38990203

RÉSUMÉ

Atherosclerosis (AS) is the main pathological basis of cardiovascular diseases such as coronary heart disease. Black phosphorus quantum dots (BPQDs) are a novel nanomaterial with good optical properties and biocompatibility, which was applied in the treatment of AS in mice, with good results shown in our previous study. In this study, BPQDs were injected into high-fat diet-fed apolipoprotein E knockout mice as a preventive drug for 12 weeks. Simvastatin, a classic preventive drug for AS, was used as a control to verify the preventive effect of BPQDs. The results showed that after preventive treatment with BPQDs, the plaque area in mice was significantly reduced, the vascular elasticity was increased, and serum lipid levels were significantly lower than those in the model group. To explore the mechanism, macrophages were induced to become foam cells using oxidized low-density lipoprotein. We found that BPQDs treatment could increase cell autophagy, thereby regulating intracellular lipid metabolism. Taken together, these data revealed that BPQDs may serve as a functional drug in preventing the development of AS.


Sujet(s)
Athérosclérose , Alimentation riche en graisse , Phosphore , Boîtes quantiques , Animaux , Alimentation riche en graisse/effets indésirables , Athérosclérose/prévention et contrôle , Souris , Phosphore/sang , Souris knockout , Apolipoprotéines E/génétique , Mâle , Autophagie/effets des médicaments et des substances chimiques , Souris invalidées pour les gènes ApoE , Métabolisme lipidique/effets des médicaments et des substances chimiques , Modèles animaux de maladie humaine , Plaque d'athérosclérose/prévention et contrôle , Lipoprotéines LDL/métabolisme , Lipoprotéines LDL/sang , Simvastatine/pharmacologie , Macrophages/effets des médicaments et des substances chimiques , Macrophages/métabolisme , Cellules spumeuses/effets des médicaments et des substances chimiques , Cellules spumeuses/métabolisme
15.
FASEB J ; 38(13): e23806, 2024 Jul 15.
Article de Anglais | MEDLINE | ID: mdl-38970404

RÉSUMÉ

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).


Sujet(s)
Ferroptose , Lipogenèse , Lipoprotéines LDL , Protéine-1 de liaison à l'élément de régulation des stérols , Sujet âgé , Femelle , Humains , Mâle , Adulte d'âge moyen , Athérosclérose/métabolisme , Athérosclérose/anatomopathologie , Maladie des artères coronaires/métabolisme , Maladie des artères coronaires/anatomopathologie , Cellules endothéliales/métabolisme , Cellules endothéliales de la veine ombilicale humaine/métabolisme , Peroxydation lipidique , Lipoprotéines LDL/métabolisme , Acyl-(acyl-carrier-protein)desaturase/métabolisme , Acyl-(acyl-carrier-protein)desaturase/génétique , Protéine-1 de liaison à l'élément de régulation des stérols/métabolisme , Protéine-1 de liaison à l'élément de régulation des stérols/génétique
16.
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
17.
Methods Mol Biol ; 2816: 223-239, 2024.
Article de Anglais | MEDLINE | ID: mdl-38977602

RÉSUMÉ

Lipoproteins in plasma are constituted by the least dense chylomicron, very-low-density lipoprotein (VLDL), low-density lipoprotein (LDL), and high-density lipoprotein (HDL) that can be separated using commercially available medium such as iodixanol. Iodixanol constitutes the self-generated density gradient to fractionate lipoproteins by rapid ultracentrifugation method, replacing time-consuming protocols. Filling the centrifuge tubes is technically easier and faster than layering salt gradients and is reproducible. The separated lipoproteins by this method are closest to the native state with 80 to 100% recovery possible. Low-density lipoprotein is the major carrier of cholesterol in systemic circulation. The plasma isolated LDL is purified to be used as native LDL and for the preparation of oxidized LDL (oxLDL). The oxLDL is characterized for its oxidation, by various methods based on assay of the lipid and protein oxidation products such as TBARS, conjugated diene formation, and by other methods such as agarose gel electrophoresis. Rapid isolation of LDL particles from human plasma is useful for lipid peroxidation studies, characterization of subclass for functional studies and clinical correlation especially in cardiovascular diseases apart from lipidomic, and proteomic studies. OxLDL preparations are done in vitro chiefly based on copper-induced oxidation; glucose and other prooxidants. Which are used for various studies using animal model and in vitro cell models especially to understand macrophage-mediated atheroma formation, vascular endothelial cell dysfunction, cell signaling studies has scope for extensive research in metabolic dysfunction of various cells.  This chapter deals with one of the applications in the in vitro cell models using macrophage (THP-1 cell line) and human retinal pigment epithelial cell (ARPE-19 cell line) to study the oxLDL uptake using fluorescently labeled oxidized LDL (DiI-oxLDL).


Sujet(s)
Lipoprotéines LDL , Lipoprotéines LDL/métabolisme , Humains , Oxydoréduction , Lignée cellulaire , Cellules THP-1 , Acides triiodo-benzoïques
18.
Methods Mol Biol ; 2816: 253-263, 2024.
Article de Anglais | MEDLINE | ID: mdl-38977604

RÉSUMÉ

Lipids are compounds involved in many biologic functions including cell structure, metabolism, energy storage and are involved in signaling. A prominent lipid in these functions is cholesterol. Cholesterol also plays a part in the signaling of melanocytes, which contain melanosomes. The maturation of these melanosomes happens during melanocyte growth. The deficit of melanogenesis or melanosome maturation is associated with ocular albinism in the eye. Aberrations of melanosome maturation are also associated with pigment dispersion syndrome. Albinism and pigment dispersion manifestations are systemic. Both melanogenesis and melanocyte maturation are affected by cholesterol metabolism. Cholesterol signaling is a part of many pathways in the body, and evaluating these signals can have implications in systemic disease processes of melanogenesis and melanosome maturation, like ocular albinism and pigment dispersion. Cholesterol is carried by lipoprotein particles. Low-density lipoprotein (LDL) is usually the transport vehicle for cholesterol to reach tissues and organelles. The LDL uptake on cells often sends out a cascade of internal signaling within the cells. We describe here LDL signaling related to lipase activity changes using enzymatic methods with a kit. We describe analyses of cholesterol esters and free cholesterol with liquid chromatography and gas chromatography with or in tandem with mass spectrometry (GC-MS and LC-MS/MS). These analyses will provide insight into melanosome maturation and melanogenesis. The methods described here are applicable to all melanocytes within the body of a model mammalian organism.


Sujet(s)
Cholestérol , Iris , Mélanocytes , Transduction du signal , Mélanocytes/métabolisme , Humains , Cholestérol/métabolisme , Iris/métabolisme , Lipoprotéines/métabolisme , Mélanosomes/métabolisme , Lipoprotéines LDL/métabolisme , Spectrométrie de masse en tandem/méthodes , Chromatographie gazeuse-spectrométrie de masse/méthodes , Chromatographie en phase liquide/méthodes , Triacylglycerol lipase/métabolisme , Mélanines/métabolisme , Cholestérol ester/métabolisme
19.
Arterioscler Thromb Vasc Biol ; 44(9): 2053-2068, 2024 Sep.
Article de Anglais | MEDLINE | ID: mdl-38989581

RÉSUMÉ

BACKGROUND: In early atherosclerosis, circulating LDLs (low-density lipoproteins) traverse individual endothelial cells by an active process termed transcytosis. The CANTOS trial (Canakinumab Antiinflammatory Thrombosis Outcome Study) treated advanced atherosclerosis using a blocking antibody for IL-1ß (interleukin-1ß); this significantly reduced cardiovascular events. However, whether IL-1ß regulates early disease, particularly LDL transcytosis, remains unknown. METHODS: We used total internal reflection fluorescence microscopy to quantify transcytosis by human coronary artery endothelial cells exposed to IL-1ß. To investigate transcytosis in vivo, we injected wild-type and knockout mice with IL-1ß and LDL to visualize acute LDL deposition in the aortic arch. RESULTS: Exposure to picomolar concentrations of IL-1ß induced transcytosis of LDL but not of albumin by human coronary artery endothelial cells. Surprisingly, expression of the 2 known receptors for LDL transcytosis, ALK-1 (activin receptor-like kinase-1) and SR-BI (scavenger receptor BI), was unchanged or decreased. Instead, IL-1ß increased the expression of the LDLR (LDL receptor); this was unexpected because LDLR is not required for LDL transcytosis. Overexpression of LDLR had no effect on basal LDL transcytosis. However, knockdown of LDLR abrogated the effect of IL-1ß on transcytosis rates while the depletion of Cav-1 (caveolin-1) did not. Since LDLR was necessary but overexpression had no effect, we reasoned that another player must be involved. Using public RNA sequencing data to curate a list of Rab (Ras-associated binding) GTPases affected by IL-1ß, we identified Rab27a. Overexpression of Rab27a alone had no effect on basal transcytosis, but its knockdown prevented induction by IL-1ß. This was phenocopied by depletion of the Rab27a effector JFC1 (synaptotagmin-like protein 1). In vivo, IL-1ß increased LDL transcytosis in the aortic arch of wild-type but not Ldlr-/- or Rab27a-deficient mice. The JFC1 inhibitor nexinhib20 also blocked IL-1ß-induced LDL accumulation in the aorta. CONCLUSIONS: IL-1ß induces LDL transcytosis by a distinct pathway requiring LDLR and Rab27a; this route differs from basal transcytosis. We speculate that induction of transcytosis by IL-1ß may contribute to the acceleration of early disease.


Sujet(s)
Vaisseaux coronaires , Cellules endothéliales , Interleukine-1 bêta , Lipoprotéines LDL , Souris knockout , Récepteurs aux lipoprotéines LDL , Transduction du signal , Transcytose , Protéines G rab , Interleukine-1 bêta/métabolisme , Animaux , Humains , Récepteurs aux lipoprotéines LDL/génétique , Récepteurs aux lipoprotéines LDL/métabolisme , Protéines G rab/métabolisme , Protéines G rab/génétique , Cellules endothéliales/métabolisme , Cellules endothéliales/effets des médicaments et des substances chimiques , Lipoprotéines LDL/métabolisme , Vaisseaux coronaires/métabolisme , Vaisseaux coronaires/effets des médicaments et des substances chimiques , Cellules cultivées , Souris de lignée C57BL , Cavéoline-1/métabolisme , Cavéoline-1/génétique , Maladies de l'aorte/métabolisme , Maladies de l'aorte/génétique , Maladies de l'aorte/anatomopathologie , Modèles animaux de maladie humaine , Aorte thoracique/métabolisme , Aorte thoracique/effets des médicaments et des substances chimiques , Athérosclérose/métabolisme , Athérosclérose/génétique , Athérosclérose/anatomopathologie , Mâle , Souris
20.
Phytomedicine ; 132: 155864, 2024 Sep.
Article de Anglais | MEDLINE | ID: mdl-39032281

RÉSUMÉ

BACKGROUND: Atherosclerosis is a long-lasting inflammatory condition affecting the walls of arteries, marked by the buildup of fats, plaque formation, and vascular remodeling. Recent findings highlight the significance of cholesterol removal pathways in influencing atherosclerosis, yet the connection between cholesterol removal and regulation of macrophage inflammation remains poorly understood. RBAP could serve as an anti-inflammatory agent; however, its role in atherosclerosis and the mechanism behind it are still not well understood. PURPOSE: The objective of this research is to explore how RBAP impacts cholesterol efflux, which is a considerable element in the advancement of atherosclerosis. METHODS: An atherosclerosis mouse model was established by using an ApoE KO strain mouse on a high-fat diet (HFD) to assess the effects of RBAP, conducted either orally or through injection. Additionally, in vitro experiments were conducted where the induction of THP-1 cells was conducted for the differentiation towards macrophages, and along with mouse RAW264.7 cells, were challenged with ox-LDL to evaluate the impact of RBAP. RESULTS: In this study, RBAP was found to reduce the production and downregulate TNF-α, IL-1ß, and IL-6 levels and inhibited the activation of the TLR4/MyD88/NF-κB signaling in atherosclerosis model mice, as well as in ox-LDL-challenged THP-1 cells and mouse RAW264.7 macrophages. RBAP's effectiveness also improved the enhancement of reverse cholesterol transport (RCT) and cholesterol removal to HDL and apoA1 by increasing the activity of genes related to cholesterol removal PPARγ/LXRα/ABCA1/ABCG1, both in ApoE-/- mice and in THP-1 cells and mouse RAW264.7 macrophages. Notably, RBAP exerted similar effects on atherosclerosis model mice and macrophages to those of TAK-242, an inhibitor of the TLR4 signaling. When RBAP and TAK-242 were applied simultaneously, the improvement was not enhanced compared with either RBAP or TAK-242 treatment alone. CONCLUSION: These findings suggest that RBAP, as a TLR4 inhibitor, has anti-atherosclerotic effects by improving inflammation and promoting cholesterol effection, indicating its therapeutic potential in intervening atherosclerosis.


Sujet(s)
Athérosclérose , Différenciation cellulaire , Cholestérol , Cellules spumeuses , Macrophages , Oryza , Récepteur de type Toll-4 , Animaux , Athérosclérose/traitement médicamenteux , Souris , Cholestérol/métabolisme , Cellules spumeuses/effets des médicaments et des substances chimiques , Cellules spumeuses/métabolisme , Cellules RAW 264.7 , Différenciation cellulaire/effets des médicaments et des substances chimiques , Humains , Récepteur de type Toll-4/métabolisme , Macrophages/effets des médicaments et des substances chimiques , Macrophages/métabolisme , Modèles animaux de maladie humaine , Cellules THP-1 , Mâle , Alimentation riche en graisse , Membre-1 de la sous-famille A des transporteurs à cassette liant l'ATP/métabolisme , Lipoprotéines LDL/métabolisme , Souris de lignée C57BL , Peptides/pharmacologie , Membre-1 de la sous-famille G des transporteurs à cassette liant l'ATP/métabolisme , Souris invalidées pour les gènes ApoE , Facteur de transcription NF-kappa B/métabolisme , Apolipoprotéines E , Anti-inflammatoires/pharmacologie
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE