Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 10.880
Filtrer
1.
Sci Rep ; 14(1): 15331, 2024 07 03.
Article de Anglais | MEDLINE | ID: mdl-38961200

RÉSUMÉ

Proprotein convertase subtilisin/kexin type 9 (PCSK9) has emerged as a promising therapeutic target to reduce lipids. In 2020, we reported a chimeric camelid-human heavy chain antibody VHH-B11-Fc targeting PCSK9. Recently, it was verified that VHH-B11 binds one linear epitope in the PCSK9 hinge region. To enhance its druggability, we have developed a novel biparatopic B11-H2-Fc Ab herein. Thereinto, surface plasmon resonance (SPR) confirmed the epitope differences in binding-PCSK9 among VHH-B11, VHH-H2 and the approved Repatha. Additionally, SPR revealed the B11-H2-Fc exhibits an avidity of approximately 0.036 nM for PCSK9, representing a considerable increase compared to VHH-B11-Fc (~ 0.69 nM). Moreover, we found the Repatha and B11-H2-Fc exhibited > 95% PCSK9 inhibition efficiency compared to approximately 48% for the VHH-Fc at 7.4 nM (P < 0.0005). Further, we verified its biological activity using the human hepatoma cells G2 model, where the B11-H2-Fc exhibited almost 100% efficiency in PCSK9 inhibition at only 0.75 µM. The immunoblotting results of low-density lipoprotein cholesterol (LDL-c) uptake assay also demonstrated the excellent performance of B11-H2-Fc on recovering the LDL-c receptor (LDLR), as strong as the Repatha (P > 0.05). These findings provide the first evidence of the efficacy of a novel Ab targeting PCSK9 in the field of lipid-lowering drugs.


Sujet(s)
Proprotéine convertase 9 , Humains , Proprotéine convertase 9/métabolisme , Proprotéine convertase 9/immunologie , Cellules HepG2 , Inhibiteurs de PCSK9 , Résonance plasmonique de surface , Récepteurs aux lipoprotéines LDL/métabolisme , Épitopes/immunologie , Lipoprotéines LDL/métabolisme , Lipoprotéines LDL/immunologie
2.
J Transl Med ; 22(1): 615, 2024 Jul 03.
Article de Anglais | MEDLINE | ID: mdl-38961500

RÉSUMÉ

OBJECTIVE: To explore the correlation between asthma risk and genetic variants affecting the expression or function of lipid-lowering drug targets. METHODS: We conducted Mendelian randomization (MR) analyses using variants in several genes associated with lipid-lowering medication targets: HMGCR (statin target), PCSK9 (alirocumab target), NPC1L1 (ezetimibe target), APOB (mipomersen target), ANGPTL3 (evinacumab target), PPARA (fenofibrate target), and APOC3 (volanesorsen target), as well as LDLR and LPL. Our objective was to investigate the relationship between lipid-lowering drugs and asthma through MR. Finally, we assessed the efficacy and stability of the MR analysis using the MR Egger and inverse variance weighted (IVW) methods. RESULTS: The elevated triglyceride (TG) levels associated with the APOC3, and LPL targets were found to increase asthma risk. Conversely, higher LDL-C levels driven by LDLR were found to decrease asthma risk. Additionally, LDL-C levels (driven by APOB, NPC1L1 and HMGCR targets) and TG levels (driven by the LPL target) were associated with improved lung function (FEV1/FVC). LDL-C levels driven by PCSK9 were associated with decreased lung function (FEV1/FVC). CONCLUSION: In conclusion, our findings suggest a likely causal relationship between asthma and lipid-lowering drugs. Moreover, there is compelling evidence indicating that lipid-lowering therapies could play a crucial role in the future management of asthma.


Sujet(s)
Asthme , Hypolipémiants , Analyse de randomisation mendélienne , Humains , Asthme/génétique , Asthme/traitement médicamenteux , Hypolipémiants/usage thérapeutique , Hypolipémiants/pharmacologie , Proprotéine convertase 9/génétique , Études d'associations génétiques , Poumon/effets des médicaments et des substances chimiques , Poumon/anatomopathologie , Lipoprotein lipase/génétique , Triglycéride/sang , Récepteurs aux lipoprotéines LDL/génétique , Hydroxymethylglutaryl-CoA reductases/génétique , Protéine-3 de type angiopoïétine , Protéines semblables à l'angiopoïétine/génétique , Apolipoprotéine C-III/génétique , Apolipoprotéines B/génétique , Tests de la fonction respiratoire , Cholestérol LDL/sang , Protéines de transport membranaire , Récepteur PPAR alpha
3.
Lipids Health Dis ; 23(1): 182, 2024 Jun 12.
Article de Anglais | MEDLINE | ID: mdl-38867270

RÉSUMÉ

BACKGROUND: Familial hypercholesterolemia (FH) is a common inherited metabolic disease that causes premature atherosclerosis, cardiovascular disease, and even death at a young age. Approximately 95% of FH-causing genetic variants that have been identified are in the LDLR gene. However, only 10% of the FH population worldwide has been diagnosed and adequately treated, due to the existence of numerous unidentified variants, uncertainties in the pathogenicity scoring of many variants, and a substantial number of individuals lacking access to genetic testing. OBJECTIVE: The aim of this study was to identify a novel variant in the LDLR gene that causes FH in a Chinese family, thereby expanding the spectrum of FH-causing variants. METHODS: Patients were recruited from Beijing Anzhen Hospital, Capital Medical University. FH diagnosis was made according to the Dutch Lipid Clinical Network (DLCN) criteria. Whole-exome sequencing (WES) was conducted to identify the FH-causing variant in the proband, and amplicon sequencing was used to verify the variant in his family members. RESULTS: A three-generation Chinese family was recruited, and two FH patients were clinically diagnosed, both without known FH-causing variants. These two FH patients and another possible patient carried a novel variant, NC_000019.9(NM_000527.5):c.89_92dup (NP_000518.1:p.Phe32Argfs*21), in the ligand-binding domain of the low-density lipoprotein (LDL) receptor that led to a frameshift. The FH adults in the family showed severe clinical symptoms and statin therapy resistance. CONCLUSION: This study identified a novel pathogenic LDLR variant, c.89_92dup, associated with severe FH clinical manifestations and statin therapy resistance.


Sujet(s)
Mutation avec décalage du cadre de lecture , Hyperlipoprotéinémie de type II , Pedigree , Récepteurs aux lipoprotéines LDL , Humains , Hyperlipoprotéinémie de type II/génétique , Hyperlipoprotéinémie de type II/diagnostic , Récepteurs aux lipoprotéines LDL/génétique , Mâle , Mutation avec décalage du cadre de lecture/génétique , Femelle , Adulte , Adulte d'âge moyen ,
4.
J Cardiovasc Pharmacol ; 84(1): 45-57, 2024 Jul 01.
Article de Anglais | MEDLINE | ID: mdl-38922585

RÉSUMÉ

ABSTRACT: Atherosclerosis (AS) is a chronic progressive disease caused by various factors and causes various cerebrovascular and cardiovascular diseases (CVDs). Reducing the plasma levels of low-density lipoprotein cholesterol is the primary goal in preventing and treating AS. Proprotein convertase subtilisin/kexin type 9 (PCSK9) plays a crucial role in regulating low-density lipoprotein cholesterol metabolism. Panax notoginseng has potent lipid-reducing effects and protects against CVDs, and its saponins induce vascular dilatation, inhibit thrombus formation, and are used in treating CVDs. However, the anti-AS effect of the secondary metabolite, 20( S )-protopanaxatriol (20( S )-PPT), remains unclear. In this study, the anti-AS effect and molecular mechanism of 20( S )-PPT were investigated in vivo and in vitro by Western blotting, real-time polymerase chain reaction, enzyme-linked immunosorbent assay, immunofluorescence staining, and other assays. The in vitro experiments revealed that 20( S )-PPT reduced the levels of PCSK9 in the supernatant of HepG2 cells, upregulated low-density lipoprotein receptor protein levels, promoted low-density lipoprotein uptake by HepG2 cells, and reduced PCSK9 mRNA transcription by upregulating the levels of forkhead box O3 protein and mRNA and decreasing the levels of HNF1α and SREBP2 protein and mRNA. The in vivo experiments revealed that 20( S )-PPT upregulated aortic α-smooth muscle actin expression, increased the stability of atherosclerotic plaques, and reduced aortic plaque formation induced by a high-cholesterol diet in ApoE -/- mice (high-cholesterol diet-fed group). Additionally, 20( S )-PPT reduced the aortic expression of CD68, reduced inflammation in the aortic root, and alleviated the hepatic lesions in the high-cholesterol diet-fed group. The study revealed that 20( S )-PPT inhibited low-density lipoprotein receptor degradation via PCSK9 to alleviate AS.


Sujet(s)
Aorte , Maladies de l'aorte , Athérosclérose , Modèles animaux de maladie humaine , Souris de lignée C57BL , Souris invalidées pour les gènes ApoE , Plaque d'athérosclérose , Proprotéine convertase 9 , Récepteurs aux lipoprotéines LDL , Sapogénines , Animaux , Athérosclérose/métabolisme , Athérosclérose/anatomopathologie , Athérosclérose/traitement médicamenteux , Athérosclérose/prévention et contrôle , Athérosclérose/génétique , Sapogénines/pharmacologie , Proprotéine convertase 9/métabolisme , Proprotéine convertase 9/génétique , Récepteurs aux lipoprotéines LDL/génétique , Récepteurs aux lipoprotéines LDL/métabolisme , Humains , Mâle , Maladies de l'aorte/anatomopathologie , Maladies de l'aorte/prévention et contrôle , Maladies de l'aorte/métabolisme , Maladies de l'aorte/génétique , Maladies de l'aorte/traitement médicamenteux , Aorte/effets des médicaments et des substances chimiques , Aorte/métabolisme , Aorte/anatomopathologie , Protéolyse/effets des médicaments et des substances chimiques , Cellules HepG2 , Inhibiteurs de PCSK9 , Transduction du signal/effets des médicaments et des substances chimiques , Protéine-2 de liaison à l'élément de régulation des stérols/métabolisme , Protéine-2 de liaison à l'élément de régulation des stérols/génétique , Souris , Alimentation riche en graisse , Apolipoprotéines E
5.
Circ Res ; 135(2): 335-349, 2024 Jul 05.
Article de Anglais | MEDLINE | ID: mdl-38828596

RÉSUMÉ

BACKGROUND: Individuals with type 1 diabetes (T1D) generally have normal or even higher HDL (high-density lipoprotein)-cholesterol levels than people without diabetes yet are at increased risk for atherosclerotic cardiovascular disease (CVD). Human HDL is a complex mixture of particles that can vary in cholesterol content by >2-fold. To investigate if specific HDL subspecies contribute to the increased atherosclerosis associated with T1D, we created mouse models of T1D that exhibit human-like HDL subspecies. We also measured HDL subspecies and their association with incident CVD in a cohort of people with T1D. METHODS: We generated LDL receptor-deficient (Ldlr-/-) mouse models of T1D expressing human APOA1 (apolipoprotein A1). Ldlr-/-APOA1Tg mice exhibited the main human HDL subspecies. We also generated Ldlr-/-APOA1Tg T1D mice expressing CETP (cholesteryl ester transfer protein), which had lower concentrations of large HDL subspecies versus mice not expressing CETP. HDL particle concentrations and sizes and proteins involved in lipoprotein metabolism were measured by calibrated differential ion mobility analysis and targeted mass spectrometry in the mouse models of T1D and in a cohort of individuals with T1D. Endothelial transcytosis was analyzed by total internal reflection fluorescence microscopy. RESULTS: Diabetic Ldlr-/-APOA1Tg mice were severely hyperglycemic and hyperlipidemic and had markedly elevated plasma APOB levels versus nondiabetic littermates but were protected from the proatherogenic effects of diabetes. Diabetic Ldlr-/-APOA1Tg mice expressing CETP lost the atheroprotective effect and had increased lesion necrotic core areas and APOB accumulation, despite having lower plasma APOB levels. The detrimental effects of low concentrations of larger HDL particles in diabetic mice expressing CETP were not explained by reduced cholesterol efflux. Instead, large HDL was more effective than small HDL in preventing endothelial transcytosis of LDL mediated by scavenger receptor class B type 1. Finally, in humans with T1D, increased concentrations of larger HDL particles relative to APOB100 negatively predicted incident CVD independently of HDL-cholesterol levels. CONCLUSIONS: Our results suggest that the balance between APOB lipoproteins and the larger HDL subspecies contributes to atherosclerosis progression and incident CVD in the setting of T1D and that larger HDLs exert atheroprotective effects on endothelial cells rather than by promoting macrophage cholesterol efflux.


Sujet(s)
Apolipoprotéine A-I , Athérosclérose , Diabète de type 1 , Récepteurs aux lipoprotéines LDL , Animaux , Athérosclérose/métabolisme , Athérosclérose/génétique , Athérosclérose/sang , Athérosclérose/anatomopathologie , Humains , Diabète de type 1/métabolisme , Diabète de type 1/sang , Souris , Récepteurs aux lipoprotéines LDL/génétique , Récepteurs aux lipoprotéines LDL/déficit , Récepteurs aux lipoprotéines LDL/métabolisme , Apolipoprotéine A-I/sang , Apolipoprotéine A-I/métabolisme , Mâle , Protéines de transfert des esters de cholestérol/génétique , Protéines de transfert des esters de cholestérol/métabolisme , Protéines de transfert des esters de cholestérol/sang , Souris knockout , Femelle , Souris de lignée C57BL , Lipoprotéines HDL/sang , Lipoprotéines HDL/métabolisme , Souris transgéniques , Apolipoprotéine B-100/métabolisme , Apolipoprotéine B-100/génétique , Apolipoprotéine B-100/sang , Adulte d'âge moyen , Modèles animaux de maladie humaine , Adulte
6.
J Am Heart Assoc ; 13(12): e034434, 2024 Jun 18.
Article de Anglais | MEDLINE | ID: mdl-38879446

RÉSUMÉ

BACKGROUND: Familial hypercholesterolemia (FH), while highly prevalent, is a significantly underdiagnosed monogenic disorder. Improved detection could reduce the large number of cardiovascular events attributable to poor case finding. We aimed to assess whether machine learning algorithms outperform clinical diagnostic criteria (signs, history, and biomarkers) and the recommended screening criteria in the United Kingdom in identifying individuals with FH-causing variants, presenting a scalable screening criteria for general populations. METHODS AND RESULTS: Analysis included UK Biobank participants with whole exome sequencing, classifying them as having FH when (likely) pathogenic variants were detected in their LDLR, APOB, or PCSK9 genes. Data were stratified into 3 data sets for (1) feature importance analysis; (2) deriving state-of-the-art statistical and machine learning models; (3) evaluating models' predictive performance against clinical diagnostic and screening criteria: Dutch Lipid Clinic Network, Simon Broome, Make Early Diagnosis to Prevent Early Death, and Familial Case Ascertainment Tool. One thousand and three of 454 710 participants were classified as having FH. A Stacking Ensemble model yielded the best predictive performance (sensitivity, 74.93%; precision, 0.61%; accuracy, 72.80%, area under the receiver operating characteristic curve, 79.12%) and outperformed clinical diagnostic criteria and the recommended screening criteria in identifying FH variant carriers within the validation data set (figures for Familial Case Ascertainment Tool, the best baseline model, were 69.55%, 0.44%, 65.43%, and 71.12%, respectively). Our model decreased the number needed to screen compared with the Familial Case Ascertainment Tool (164 versus 227). CONCLUSIONS: Our machine learning-derived model provides a higher pretest probability of identifying individuals with a molecular diagnosis of FH compared with current approaches. This provides a promising, cost-effective scalable tool for implementation into electronic health records to prioritize potential FH cases for genetic confirmation.


Sujet(s)
Apolipoprotéine B-100 , Hyperlipoprotéinémie de type II , Apprentissage machine , Proprotéine convertase 9 , Humains , Hyperlipoprotéinémie de type II/génétique , Hyperlipoprotéinémie de type II/diagnostic , Hyperlipoprotéinémie de type II/épidémiologie , Femelle , Mâle , Proprotéine convertase 9/génétique , Apolipoprotéine B-100/génétique , Adulte d'âge moyen , Récepteurs aux lipoprotéines LDL/génétique , Royaume-Uni/épidémiologie , , Dépistage génétique/méthodes , Adulte , Valeur prédictive des tests , Prédisposition génétique à une maladie , Mutation
7.
Cell Rep Med ; 5(6): 101614, 2024 Jun 18.
Article de Anglais | MEDLINE | ID: mdl-38897173

RÉSUMÉ

Proprotein convertase subtilisin/kexin type 9 (PCSK9) binds to the low-density lipoprotein receptor (LDLR) and mediates its internalization and degradation, resulting in an increase in LDL cholesterol levels. Recently, PCSK9 emerged as a therapeutic target for hypercholesterolemia and atherosclerosis. In this study, we develop a PCSK9 nanoparticle (NP) vaccine by covalently conjugating the catalytic domain (aa 153-aa 454, D374Y) of PCSK9 to self-assembled 24-mer ferritin NPs. We demonstrate that the PCSK9 NP vaccine effectively induces interfering antibodies against PCSK9 and reduces serum lipids levels in both a high-fat diet-induced hypercholesterolemia model and an adeno-associated virus-hPCSK9D374Y-induced hypercholesterolemia model. Additionally, the vaccine significantly reduces plaque lesion areas in the aorta and macrophages infiltration in an atherosclerosis mouse model. Furthermore, we discover that the vaccine's efficacy relied on T follicular help cells and LDLR. Overall, these findings suggest that the PCSK9 NP vaccine holds promise as an effective treatment for hypercholesterolemia and atherosclerosis.


Sujet(s)
Athérosclérose , Modèles animaux de maladie humaine , Hypercholestérolémie , Nanoparticules , Proprotéine convertase 9 , Récepteurs aux lipoprotéines LDL , Vaccins , Proprotéine convertase 9/immunologie , Proprotéine convertase 9/métabolisme , Animaux , Hypercholestérolémie/anatomopathologie , Nanoparticules/composition chimique , Vaccins/immunologie , Souris , Récepteurs aux lipoprotéines LDL/métabolisme , Athérosclérose/prévention et contrôle , Athérosclérose/immunologie , Athérosclérose/anatomopathologie , Souris de lignée C57BL , Humains , Alimentation riche en graisse , Mâle ,
8.
Clin Transl Sci ; 17(6): e13836, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38845393

RÉSUMÉ

Homozygous familial hypercholesterolemia (HoFH) is a rare and serious genetic condition characterized by premature cardiovascular disease due to severely elevated low-density lipoprotein cholesterol (LDL-C). HoFH primarily results from loss-of-function (LOF) mutations in the LDL receptor (LDLR), reducing LDL-C clearance such that patients experience severe hypercholesterolemia, exacerbating the risk of developing cardiovascular events. Treatment options such as statins, lomitapide, ezetimibe, proprotein convertase subtilisin/kexin type 9 inhibitors, and apheresis help lower LDL-C; however, many patients with HoFH still fail to reach their target LDL-C levels and many of these lipid-lowering therapies are not indicated for pediatric use. Angiopoietin-like protein 3 (ANGPTL3) has been identified as a target to treat elevated LDL-C by acting as a natural inhibitor of lipoprotein lipase (LPL) and endothelial lipase (EL), enzymes involved in the hydrolysis of the triglyceride and phospholipid content of very low-density lipoproteins. Persons heterozygous for LOF mutations in ANGPTL3 were reported to have lower LDL-C than non-carriers and lower risk of coronary artery disease. Evinacumab is a first-in-class human monoclonal antibody that specifically binds to ANGPTL3 to prevent its inhibition of LPL and EL. In clinical trials, a 15 mg/kg intravenous dose every 4 weeks has shown a mean percent change from baseline in LDL-C of ~50% in adult, adolescent, and pediatric patients with HoFH. This mini review article describes the mechanism of action of evinacumab, evinacumab population PK and PD modeling, and clinical development history of evinacumab for the treatment of HoFH.


Sujet(s)
Hyperlipoprotéinémie de type II , , Humains , Hyperlipoprotéinémie de type II/traitement médicamenteux , Hyperlipoprotéinémie de type II/génétique , Hyperlipoprotéinémie de type II/sang , Protéine-3 de type angiopoïétine , Cholestérol LDL/sang , Anticorps monoclonaux/usage thérapeutique , Anticorps monoclonaux/pharmacologie , Anticorps neutralisants à large spectre , Animaux , Anticholestérolémiants/usage thérapeutique , Anticholestérolémiants/pharmacologie , Anticholestérolémiants/administration et posologie , Récepteurs aux lipoprotéines LDL/métabolisme , Récepteurs aux lipoprotéines LDL/génétique
9.
Cell Death Dis ; 15(6): 389, 2024 Jun 03.
Article de Anglais | MEDLINE | ID: mdl-38830896

RÉSUMÉ

Apolipoprotein O (APOO) plays a critical intracellular role in regulating lipid metabolism. Here, we investigated the roles of APOO in metabolism and atherogenesis in mice. Hepatic APOO expression was increased in response to hyperlipidemia but was inhibited after simvastatin treatment. Using a novel APOO global knockout (Apoo-/-) model, it was found that APOO depletion aggravated diet-induced obesity and elevated plasma cholesterol levels. Upon crossing with low-density lipoprotein receptor (LDLR) and apolipoprotein E (APOE) knockout hyperlipidemic mouse models, Apoo-/- Apoe-/- and Apoo-/- Ldlr-/- mice exhibited elevated plasma cholesterol levels, with more severe atherosclerotic lesions than littermate controls. This indicated the effects of APOO on cholesterol metabolism independent of LDLR and APOE. Moreover, APOO deficiency reduced cholesterol excretion through bile and feces while decreasing phospholipid unsaturation by inhibiting NRF2 and CYB5R3. Restoration of CYB5R3 expression in vivo by adeno-associated virus (AAV) injection reversed the reduced degree of phospholipid unsaturation while decreasing blood cholesterol levels. This represents the first in vivo experimental validation of the role of APOO in plasma cholesterol metabolism independent of LDLR and elucidates a previously unrecognized cholesterol metabolism pathway involving NRF2/CYB5R3. APOO may be a metabolic regulator of total-body cholesterol homeostasis and a target for atherosclerosis management. Apolipoprotein O (APOO) regulates plasma cholesterol levels and atherosclerosis through a pathway involving CYB5R3 that regulates biliary and fecal cholesterol excretion, independently of the LDL receptor. In addition, down-regulation of APOO may lead to impaired mitochondrial function, which in turn aggravates diet-induced obesity and fat accumulation.


Sujet(s)
Cholestérol , Facteur-2 apparenté à NF-E2 , Récepteurs aux lipoprotéines LDL , Animaux , Récepteurs aux lipoprotéines LDL/métabolisme , Cholestérol/métabolisme , Facteur-2 apparenté à NF-E2/métabolisme , Souris , Souris knockout , Souris de lignée C57BL , Métabolisme lipidique , Mâle , Athérosclérose/métabolisme , Apolipoprotéines/métabolisme , Apolipoprotéines/génétique , Humains , Foie/métabolisme , Apolipoprotéines E/métabolisme , Hyperlipidémies/métabolisme
10.
Mol Biol Rep ; 51(1): 752, 2024 Jun 14.
Article de Anglais | MEDLINE | ID: mdl-38874786

RÉSUMÉ

BACKGROUND: Many studies have focused on the significance of lipid regulatory genes in the pathophysiology of Coronary artery disease (CAD). ApoB XbaI (rs693) and EcoRI (rs1042031) single nucleoid polymorphisms (SNPs) were investigated to detect whether they are risk factors for CAD. Till now, this association remains uncertain. SMARCA4 (rs1122608) SNP has directly related to dyslipidemia. Loss of function mutations (LOF) in PCSK9 result in a reduction in LDL cholesterol and are associated with protection from the development of CAD. METHODS: This study was conducted on 54 CAD patients who were admitted at Internal Medicine Specialized Hospital (Cardiology Department) and 47 healthy controls. Peripheral blood samples were taken from both groups. DNA was extracted from EDTA-blood samples, then PCR- RFLP for ApoB XbaI (rs693) and EcoRI (rs1042031), SMARCA4 (rs1122608) and PCSK9 (rs505151) SNPs was done. RESULTS: No statistically significant difference was found between patients and controls as regard EcoRI SNP. XbaI (rs693) X + X + genotype was significantly higher in control group (P = 0.0355). SMARCA4 (TT, GT + TT) genotypes, and T allele (P < 0.001); PCSK9 AG genotype and G allele (P = 0.027 and 0.032 respectively) were more frequent in CAD patients than controls. CONCLUSION: SMARCA4 (rs1122608) and PCSK9 (rs505151) SNPs are significantly accompanying with the risk of CAD development in the Egyptian population. X + X + genotype appeared to have a protective effect against CAD. However, no observed association between EcoRI (rs1042031) and the risk of CAD development was found.


Sujet(s)
Maladie des artères coronaires , Prédisposition génétique à une maladie , Polymorphisme de nucléotide simple , Proprotéine convertase 9 , Récepteurs aux lipoprotéines LDL , Humains , Maladie des artères coronaires/génétique , Proprotéine convertase 9/génétique , Polymorphisme de nucléotide simple/génétique , Égypte/épidémiologie , Femelle , Mâle , Adulte d'âge moyen , Récepteurs aux lipoprotéines LDL/génétique , Études cas-témoins , Apolipoprotéines B/génétique , Facteurs de risque , Sujet âgé , Génotype , Études d'associations génétiques , Adulte , Fréquence d'allèle/génétique , Allèles , Nord-Africains , Apolipoprotéine B-100
11.
Nat Commun ; 15(1): 4906, 2024 Jun 08.
Article de Anglais | MEDLINE | ID: mdl-38851803

RÉSUMÉ

Various low-density lipoprotein receptors (LPRs) have been identified as entry factors for alphaviruses, and structures of the corresponding virion-receptor complexes have been determined. Here, we analyze the similarities and differences in the receptor binding modes of multiple alphaviruses to understand their ability to infect a wide range of hosts. We further discuss the challenges associated with the development of broad-spectrum treatment strategies against a diverse range of alphaviruses.


Sujet(s)
Alphavirus , Antiviraux , Récepteurs aux lipoprotéines LDL , Pénétration virale , Animaux , Humains , Alphavirus/effets des médicaments et des substances chimiques , Alphavirus/physiologie , Alphavirus/génétique , Infections à alphavirus/traitement médicamenteux , Infections à alphavirus/virologie , Antiviraux/usage thérapeutique , Antiviraux/pharmacologie , Liaison aux protéines , Récepteurs aux lipoprotéines LDL/métabolisme , Récepteurs aux lipoprotéines LDL/génétique , Récepteurs viraux/métabolisme , Récepteurs viraux/composition chimique , Virion/métabolisme , Pénétration virale/effets des médicaments et des substances chimiques
12.
Stem Cell Res ; 78: 103463, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-38852422

RÉSUMÉ

Familial hypercholesterolemia (FH) is a genetic disorder affecting the metabolism of lipoprotein, characterized by elevated levels of plasma concentrations of low-density lipoprotein cholesterol (LDLC). The most common FH cause is mutations within the gene that encodes for the LDL receptor (LDLR) protein. Two induced pluripotent stem cell (iPSC) lines were generated from patients with FH, each carrying a single heterozygous mutation in the LDLR gene, one is a missense mutation, c.631C > T, and the other is a splice-site mutation, c.313 + 1G > A. Both iPSC lines exhibited strong expression of pluripotency markers, demonstrated the ability to differentiate into derivatives of the three germ layers, and maintained normal karyotypes. These derived iPSC lines represent powerful tools for in vitro modeling FH and offer a promising platform for therapeutic development.


Sujet(s)
Hétérozygote , Hyperlipoprotéinémie de type II , Cellules souches pluripotentes induites , Mutation , Récepteurs aux lipoprotéines LDL , Cellules souches pluripotentes induites/métabolisme , Récepteurs aux lipoprotéines LDL/génétique , Récepteurs aux lipoprotéines LDL/métabolisme , Humains , Hyperlipoprotéinémie de type II/génétique , Hyperlipoprotéinémie de type II/métabolisme , Lignée cellulaire , Mâle , Femelle , Différenciation cellulaire
13.
Zhonghua Zhong Liu Za Zhi ; 46(5): 399-408, 2024 May 23.
Article de Chinois | MEDLINE | ID: mdl-38742353

RÉSUMÉ

Objectives: To investigate the effect of the expression of low-density lipoprotein receptor associated protein (LDLR) on the vascular abnormalities in hepatocellular carcinoma (HCC) and its mechanisms. Methods: Based on the information of Oncomine Cancer GeneChip database, we analyzed the correlation between the expression level of LDLR and the expression level of carcinoembryonic antigen (CEA) and CD31 in hepatocellular carcinoma tissues. Lentiviral transfection of short hairpin RNA target genes was used to construct LDLR-knockdown MHCC-97H and HLE hepatocellular carcinoma cells. The differential genes and their expression level changes in LDLR-knockdown hepatocellular carcinoma cells were detected by transcriptome sequencing, real-time fluorescence quantitative polymerase chain reaction, and protein immunoblotting. The gene-related signaling pathways that involve LDLR were clarified by enrichment analysis. The effect of LDLR on CEA was assessed by the detection of CEA content in conditioned medium of hepatocellular carcinoma cells. Angiogenesis assay was used to detect the effect of LDLR on the angiogenic capacity of human umbilical vein endothelial cells, as well as the role of CEA in the regulation of angiogenesis by LDLR. Immunohistochemical staining was used to detect the expression levels of LDLR in 176 hepatocellular carcinoma tissues, and CEA and CD31 in 146 hepatocellular carcinoma tissues, and analyze the correlations between the expression levels of LDLR, CEA, and CD31 in the tissues, serum CEA, and alanine transaminase (ALT). Results: Oncomine database analysis showed that the expressions of LDLR and CEA in the tissues of hepatocellular carcinoma patients with portal vein metastasis were negatively correlated (r=-0.64, P=0.001), whereas the expressions of CEA and CD31 in these tissues were positively correlated ( r=0.46, P=0.010). The transcriptome sequencing results showed that there were a total of 1 032 differentially expressed genes in the LDLR-knockdown group and the control group of MHCC-97H cells, of which 517 genes were up-regulated and 515 genes were down-regulated. The transcript expression level of CEACAM5 was significantly up-regulated in the cells of the LDLR-knockdown group. The Gene Ontology (GO) function enrichment analysis showed that the differential genes were most obviously enriched in the angiogenesis function. The Kyoto Encyclopedia of Genes and Genomes (KEGG) signaling pathway enrichment analysis showed that the relevant pathways involved mainly included the cellular adhesion patch, the extracellular matrix receptor interactions, and the interactions with the extracellular matrix receptors. The CEA content in the conditioned medium of the LDLR-knockdown group was 43.75±8.43, which was higher than that of the control group (1.15±0.14, P<0.001). The results of angiogenesis experiments showed that at 5 h, the number of main junctions, the number of main segments, and the total area of the lattice formed by HUVEC cells cultured with the conditioned medium of MHCC-97H cells in the LDLR-knockdown group were 295.3±26.4, 552.5±63.8, and 2 239 781.0±13 8211.9 square pixels, which were higher than those of the control group (113.3±23.5, 194.8±36.5, and 660 621.0±280 328.3 square pixels, respectively, all P<0.01).The number of vascular major junctions, the number of major segments, and the total area of the lattice formed by HUVEC cells cultured in conditioned medium with HLE cells in the LDLR-knockdown group were 245.3±42.4, 257.5±20.4, and 2 535 754.5±249 094.2 square pixels, respectively, which were all higher than those of the control group (113.3±23.5, 114.3±12.2, and 1 565 456.5±219 259.7 square pixels, respectively, all P<0.01). In the conditioned medium for the control group of MHCC-97H cells,the number of main junctions, the number of main segments, and the total area of the lattice formed by the addition of CEA to cultured HUVEC cells were 178.9±12.0, 286.9±12.3, and 1 966 990.0±126 249.5 spixels, which were higher than those in the control group (119.7±22.1, 202.7±33.7, and 1 421 191.0±189 837.8 square pixels, respectively). The expression of LDLR in hepatocellular carcinoma tissues was not correlated with the expression of CEA, but was negatively correlated with the expression of CD31 (r=-0.167, P=0.044), the level of serum CEA (r=-0.061, P=0.032), and the level of serum ALT(r=-0.147,P=0.05). The expression of CEA in hepatocellular carcinoma tissues was positively correlated with the expression of CD31 (r=0.192, P=0.020). The level of serum CEA was positively correlated with the level of serum ALT (r=0.164, P=0.029). Conclusion: Knocking down LDLR can promote vascular abnormalities in HCC by releasing CEA.


Sujet(s)
Carcinome hépatocellulaire , Tumeurs du foie , Néovascularisation pathologique , Récepteurs aux lipoprotéines LDL , Humains , Tumeurs du foie/métabolisme , Tumeurs du foie/génétique , Carcinome hépatocellulaire/métabolisme , Carcinome hépatocellulaire/génétique , Carcinome hépatocellulaire/anatomopathologie , Carcinome hépatocellulaire/vascularisation , Récepteurs aux lipoprotéines LDL/métabolisme , Récepteurs aux lipoprotéines LDL/génétique , Lignée cellulaire tumorale , Néovascularisation pathologique/métabolisme , Antigène carcinoembryonnaire/métabolisme , Antigène carcinoembryonnaire/génétique , Cellules endothéliales de la veine ombilicale humaine/métabolisme , Transduction du signal , Régulation de l'expression des gènes tumoraux , Techniques de knock-down de gènes , Transcriptome , Antigènes CD31/métabolisme , Antigènes CD31/génétique
14.
Arterioscler Thromb Vasc Biol ; 44(7): 1646-1657, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38695172

RÉSUMÉ

BACKGROUND: Trem2 (triggering receptor on myeloid cells 2), a surface lipid receptor, is expressed on foamy macrophages within atherosclerotic lesions and regulates cell survival, proliferation, and anti-inflammatory responses. Studies examining the role of Trem2 in atherosclerosis have shown that deletion of Trem2 leads to impaired foamy macrophage lipid uptake, proliferation, survival, and cholesterol efflux. Thus, we tested the hypothesis that administration of a Trem2 agonist antibody (AL002a) to atherogenic mice would enhance macrophage survival and decrease necrotic core formation to improve plaque stability. METHODS: To model a therapeutic intervention approach, atherosclerosis-prone mice (Ldlr [low-density lipoprotein receptor]-/-) were fed a high-fat diet for 8 weeks, then transitioned to treatment with AL002a or isotype control for an additional 8 weeks while continuing on a high-fat diet. RESULTS: AL002a-treated mice had increased lesion size in both the aortic root and whole mount aorta, which correlated with an expansion of plaque macrophage area. This expansion was due to increased macrophage survival and proliferation in plaques. Importantly, plaques from AL002a-treated mice showed improved features of plaque stability, including smaller necrotic cores, increased fibrous caps, and greater collagen deposition. Single-cell RNA sequencing of whole aorta suspensions from isotype- and AL002a-treated atherosclerotic mice revealed that Trem2 agonism dramatically altered foamy macrophage transcriptome. This included upregulation of oxidative phosphorylation and increased expression of collagen genes. In vitro studies validated that Trem2 agonism with AL002a promoted foamy macrophage oxidized low-density lipoprotein uptake, survival, and cholesterol efflux. CONCLUSIONS: Trem2 agonism expands atherosclerotic plaque macrophages by promoting cell survival and proliferation but improves features of plaque stability by rewiring foamy macrophage function to enhance cholesterol efflux and collagen deposition.


Sujet(s)
Athérosclérose , Modèles animaux de maladie humaine , Cellules spumeuses , Glycoprotéines membranaires , Souris de lignée C57BL , Souris knockout , Plaque d'athérosclérose , Récepteurs immunologiques , Animaux , Récepteurs immunologiques/agonistes , Récepteurs immunologiques/métabolisme , Récepteurs immunologiques/génétique , Glycoprotéines membranaires/agonistes , Glycoprotéines membranaires/métabolisme , Glycoprotéines membranaires/génétique , Souris , Athérosclérose/anatomopathologie , Athérosclérose/métabolisme , Athérosclérose/génétique , Athérosclérose/traitement médicamenteux , Athérosclérose/prévention et contrôle , Cellules spumeuses/métabolisme , Cellules spumeuses/anatomopathologie , Cellules spumeuses/effets des médicaments et des substances chimiques , Mâle , Récepteurs aux lipoprotéines LDL/génétique , Récepteurs aux lipoprotéines LDL/métabolisme , Récepteurs aux lipoprotéines LDL/déficit , Prolifération cellulaire/effets des médicaments et des substances chimiques , Alimentation riche en graisse , Survie cellulaire/effets des médicaments et des substances chimiques , Nécrose , Maladies de l'aorte/anatomopathologie , Maladies de l'aorte/génétique , Maladies de l'aorte/métabolisme , Maladies de l'aorte/prévention et contrôle
15.
Cell Commun Signal ; 22(1): 297, 2024 May 28.
Article de Anglais | MEDLINE | ID: mdl-38807218

RÉSUMÉ

BACKGROUND: Endoplasmic reticulum (ER) stress-mediated increases in the hepatic levels of the very low-density lipoprotein (VLDL) receptor (VLDLR) promote hepatic steatosis by increasing the delivery of triglyceride-rich lipoproteins to the liver. Here, we examined whether the NAD(+)-dependent deacetylase sirtuin 1 (SIRT1) regulates hepatic lipid accumulation by modulating VLDLR levels and the subsequent uptake of triglyceride-rich lipoproteins. METHODS: Rats fed with fructose in drinking water, Sirt1-/- mice, mice treated with the ER stressor tunicamycin with or without a SIRT1 activator, and human Huh-7 hepatoma cells transfected with siRNA or exposed to tunicamycin or different inhibitors were used. RESULTS: Hepatic SIRT1 protein levels were reduced, while those of VLDLR were upregulated in the rat model of metabolic dysfunction-associated steatotic liver disease (MASLD) induced by fructose-drinking water. Moreover, Sirt1-/- mice displayed increased hepatic VLDLR levels that were not associated with ER stress, but were accompanied by an increased expression of hypoxia-inducible factor 1α (HIF-1α)-target genes. The pharmacological inhibition or gene knockdown of SIRT1 upregulated VLDLR protein levels in the human Huh-7 hepatoma cell line, with this increase abolished by the pharmacological inhibition of HIF-1α. Finally, SIRT1 activation prevented the increase in hepatic VLDLR protein levels in mice treated with the ER stressor tunicamycin. CONCLUSIONS: Overall, these findings suggest that SIRT1 attenuates fatty liver development by modulating hepatic VLDLR levels.


Sujet(s)
Foie , Récepteurs aux lipoprotéines LDL , Sirtuine-1 , Animaux , Sirtuine-1/métabolisme , Sirtuine-1/génétique , Humains , Foie/métabolisme , Foie/effets des médicaments et des substances chimiques , Récepteurs aux lipoprotéines LDL/métabolisme , Récepteurs aux lipoprotéines LDL/génétique , Souris , Mâle , Stress du réticulum endoplasmique/effets des médicaments et des substances chimiques , Rats , Lignée cellulaire tumorale , Souris knockout , Stéatose hépatique/métabolisme , Stéatose hépatique/génétique , Stéatose hépatique/anatomopathologie , Souris de lignée C57BL , Tunicamycine/pharmacologie , Sous-unité alpha du facteur-1 induit par l'hypoxie/métabolisme , Sous-unité alpha du facteur-1 induit par l'hypoxie/génétique , Rat Sprague-Dawley
16.
Genome Med ; 16(1): 70, 2024 05 20.
Article de Anglais | MEDLINE | ID: mdl-38769532

RÉSUMÉ

BACKGROUND: Rare oncogenic driver events, particularly affecting the expression or splicing of driver genes, are suspected to substantially contribute to the large heterogeneity of hematologic malignancies. However, their identification remains challenging. METHODS: To address this issue, we generated the largest dataset to date of matched whole genome sequencing and total RNA sequencing of hematologic malignancies from 3760 patients spanning 24 disease entities. Taking advantage of our dataset size, we focused on discovering rare regulatory aberrations. Therefore, we called expression and splicing outliers using an extension of the workflow DROP (Detection of RNA Outliers Pipeline) and AbSplice, a variant effect predictor that identifies genetic variants causing aberrant splicing. We next trained a machine learning model integrating these results to prioritize new candidate disease-specific driver genes. RESULTS: We found a median of seven expression outlier genes, two splicing outlier genes, and two rare splice-affecting variants per sample. Each category showed significant enrichment for already well-characterized driver genes, with odds ratios exceeding three among genes called in more than five samples. On held-out data, our integrative modeling significantly outperformed modeling based solely on genomic data and revealed promising novel candidate driver genes. Remarkably, we found a truncated form of the low density lipoprotein receptor LRP1B transcript to be aberrantly overexpressed in about half of hairy cell leukemia variant (HCL-V) samples and, to a lesser extent, in closely related B-cell neoplasms. This observation, which was confirmed in an independent cohort, suggests LRP1B as a novel marker for a HCL-V subclass and a yet unreported functional role of LRP1B within these rare entities. CONCLUSIONS: Altogether, our census of expression and splicing outliers for 24 hematologic malignancy entities and the companion computational workflow constitute unique resources to deepen our understanding of rare oncogenic events in hematologic cancers.


Sujet(s)
Tumeurs hématologiques , Transcriptome , Humains , Tumeurs hématologiques/génétique , Épissage des ARN , Régulation de l'expression des gènes tumoraux , Oncogènes , Analyse de profil d'expression de gènes , Récepteurs aux lipoprotéines LDL/génétique
17.
J Intern Med ; 296(1): 39-52, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38704820

RÉSUMÉ

Platelet hyperreactivity and hyperlipidaemia contribute significantly to atherosclerosis. Thus, it is desirable to review the platelet-hyperlipidaemia interplay and its impact on atherogenesis. Native low-density lipoprotein (nLDL) and oxidized LDL (oxLDL) are the key proatherosclerotic components of hyperlipidaemia. nLDL binds to the platelet-specific LDL receptor (LDLR) ApoE-R2', whereas oxLDL binds to the platelet-expressed scavenger receptor CD36, lectin-type oxidized LDLR 1 and scavenger receptor class A 1. Ligation of nLDL/oxLDL induces mild platelet activation and may prime platelets for other platelet agonists. Platelets, in turn, can modulate lipoprotein metabolisms. Platelets contribute to LDL oxidation by enhancing the production of reactive oxygen species and LDLR degradation via proprotein convertase subtilisin/kexin type 9 release. Platelet-released platelet factor 4 and transforming growth factor ß modulate LDL uptake and foam cell formation. Thus, platelet dysfunction and hyperlipidaemia work in concert to aggravate atherogenesis. Hypolipidemic drugs modulate platelet function, whereas antiplatelet drugs influence lipid metabolism. The research prospects of the platelet-hyperlipidaemia interplay in atherosclerosis are also discussed.


Sujet(s)
Athérosclérose , Plaquettes , Hyperlipidémies , Lipoprotéines LDL , Humains , Athérosclérose/étiologie , Plaquettes/métabolisme , Lipoprotéines LDL/métabolisme , Activation plaquettaire/physiologie , Récepteurs aux lipoprotéines LDL/métabolisme , Hypolipémiants/usage thérapeutique
18.
Molecules ; 29(9)2024 Apr 26.
Article de Anglais | MEDLINE | ID: mdl-38731489

RÉSUMÉ

Gallic acid (GA) is a type of polyphenolic compound that can be found in a range of fruits, vegetables, and tea. Although it has been confirmed it improves non-alcoholic fatty liver disease (NAFLD), it is still unknown whether GA can improve the occurrence of NAFLD by increasing the low-density lipoprotein receptor (LDLR) accumulation and alleviating cholesterol metabolism disorders. Therefore, the present study explored the effect of GA on LDLR and its mechanism of action. The findings indicated that the increase in LDLR accumulation in HepG2 cells induced by GA was associated with the stimulation of the epidermal growth factor receptor-extracellular regulated protein kinase (EGFR-ERK1/2) signaling pathway. When the pathway was inhibited by EGFR mab cetuximab, it was observed that the activation of the EGFR-ERK1/2 signaling pathway induced by GA was also blocked. At the same time, the accumulation of LDLR protein and the uptake of LDL were also suppressed. Additionally, GA can also promote the accumulation of forkhead box O3 (FOXO3) and suppress the accumulation of hepatocyte nuclear factor-1α (HNF1α), leading to the inhibition of proprotein convertase subtilisin/kexin 9 (PCSK9) mRNA expression and protein accumulation. This ultimately results in increased LDLR protein accumulation and enhanced uptake of LDL in cells. In summary, the present study revealed the potential mechanism of GA's role in ameliorating NAFLD, with a view of providing a theoretical basis for the dietary supplementation of GA.


Sujet(s)
Acide gallique , Lipoprotéines LDL , Récepteurs aux lipoprotéines LDL , Humains , Acide gallique/pharmacologie , Récepteurs aux lipoprotéines LDL/métabolisme , Cellules HepG2 , Lipoprotéines LDL/métabolisme , Récepteurs ErbB/métabolisme , Système de signalisation des MAP kinases/effets des médicaments et des substances chimiques , Transduction du signal/effets des médicaments et des substances chimiques , Proprotéine convertase 9/métabolisme , Proprotéine convertase 9/génétique
19.
Lipids Health Dis ; 23(1): 156, 2024 May 25.
Article de Anglais | MEDLINE | ID: mdl-38796450

RÉSUMÉ

The degradation of low-density lipoprotein receptor (LDLR) is induced by proprotein convertase subtilisin/kexin type 9 (PCSK9), resulting in elevated plasma concentrations of LDL cholesterol. Therefore, inhibiting the interactions between PCSK9 and LDLR is a desirable therapeutic goal for managing hypercholesterolemia. Aptamers, which are RNA or single-stranded DNA sequences, can recognize their targets based on their secondary structure. Aptamers exhibit high selectivity and affinity for binding to target molecules. The systematic evolution of ligands by exponential enrichment (SELEX), a combination of biological approaches, is used to screen most aptamers in vitro. Due to their unique advantages, aptamers have garnered significant interest since their discovery and have found extensive applications in various fields. Aptamers have been increasingly utilized in the development of biosensors for sensitive detection of pathogens, analytes, toxins, drug residues, and malignant cells. Furthermore, similar to monoclonal antibodies, aptamers can serve as therapeutic tools. Unlike certain protein therapeutics, aptamers do not elicit antibody responses, and their modified sugars at the 2'-positions generally prevent toll-like receptor-mediated innate immune responses. The focus of this review is on aptamer-based targeting of PCSK9 and the application of aptamers both as biosensors and therapeutic agents.


Sujet(s)
Aptamères nucléotidiques , Techniques de biocapteur , Métabolisme lipidique , Proprotéine convertase 9 , Proprotéine convertase 9/métabolisme , Proprotéine convertase 9/génétique , Proprotéine convertase 9/sang , Humains , Techniques de biocapteur/méthodes , Récepteurs aux lipoprotéines LDL/métabolisme , Technique SELEX , Hypercholestérolémie/traitement médicamenteux , Hypercholestérolémie/diagnostic , Hypercholestérolémie/sang , Animaux , Inhibiteurs de PCSK9
20.
Am J Physiol Heart Circ Physiol ; 327(1): H56-H66, 2024 Jul 01.
Article de Anglais | MEDLINE | ID: mdl-38758128

RÉSUMÉ

Mouse models of congenital aortic valve malformations are useful for studying disease pathobiology, but most models have incomplete penetrance [e.g., ∼2 to 77% prevalence of bicuspid aortic valves (BAVs) across multiple models]. For longitudinal studies of pathologies associated with BAVs and other congenital valve malformations, which manifest over months in mice, it is operationally inefficient, economically burdensome, and ethically challenging to enroll large numbers of mice in studies without first identifying those with valvular abnormalities. To address this need, we established and validated a novel in vivo high-frequency (30 MHz) ultrasound imaging protocol capable of detecting aortic valvular malformations in juvenile mice. Fifty natriuretic peptide receptor 2 heterozygous mice on a low-density lipoprotein receptor-deficient background (Npr2+/-;Ldlr-/-; 32 males and 18 females) were imaged at 4 and 8 wk of age. Fourteen percent of the Npr2+/-;Ldlr-/- mice exhibited features associated with aortic valve malformations, including 1) abnormal transaortic flow patterns on color Doppler (recirculation and regurgitation), 2) peak systolic flow velocities distal to the aortic valves reaching or surpassing ∼1,250 mm/s by pulsed-wave Doppler, and 3) putative fusion of cusps along commissures and abnormal movement elucidated by two-dimensional (2-D) imaging with ultrahigh temporal resolution. Valves with these features were confirmed by ex vivo gross anatomy and histological visualization to have thickened cusps, partial fusions, or Sievers type-0 bicuspid valves. This ultrasound imaging protocol will enable efficient, cost effective, and humane implementation of studies of congenital aortic valvular abnormalities and associated pathologies in a wide range of mouse models.NEW & NOTEWORTHY We developed a high-frequency ultrasound imaging protocol for diagnosing congenital aortic valve structural abnormalities in 4-wk-old mice. Our protocol defines specific criteria to distinguish mice with abnormal aortic valves from those with normal tricuspid valves using color Doppler, pulsed-wave Doppler, and two-dimensional (2-D) imaging with ultrahigh temporal resolution. This approach enables early identification of valvular abnormalities for efficient and ethical experimental design of longitudinal studies of congenital valve diseases and associated pathologies in mice.


Sujet(s)
Valve aortique , Modèles animaux de maladie humaine , Récepteur facteur natriurétique auriculaire , Animaux , Valve aortique/malformations , Valve aortique/imagerie diagnostique , Valve aortique/anatomopathologie , Femelle , Mâle , Récepteur facteur natriurétique auriculaire/génétique , Récepteur facteur natriurétique auriculaire/déficit , Récepteur facteur natriurétique auriculaire/métabolisme , Souris , Souris knockout , Récepteurs aux lipoprotéines LDL/génétique , Récepteurs aux lipoprotéines LDL/déficit , Souris de lignée C57BL , Maladie de la valve aortique bicuspide/imagerie diagnostique
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...