Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 127
Filtrer
1.
J Clin Lipidol ; 18(3): e374-e383, 2024.
Article de Anglais | MEDLINE | ID: mdl-38782655

RÉSUMÉ

Increased cholesterol-rich, low-density, non-calcified atheromas as assessed by computer coronary tomography angiography analyses have been shown to predict myocardial infarction significantly better than coronary artery calcium score or the presence of obstructive coronary artery disease (CAD) as evaluated with standard coronary angiography. Low serum high-density lipoprotein (HDL) cholesterol values are an independent risk factor for CAD. Very small, lipid-poor preß-1 HDL particles have been shown to be most effective in promoting cellular cholesterol efflux. HDL infusions have been documented to reduce aortic atherosclerosis in cholesterol-fed animal models. However, human studies using infusions of either the HDL mimetic containing recombinant apolipoprotein (apo) A-I Milano or Cerenis Compound-001 with native recombinant apoA-I have been mainly negative in promoting coronary atherosclerosis progression as assessed by intravascular ultrasound. In contrast, a study using 7 weekly infusions of autologous delipidated HDL in six homozygous familial hypercholesterolemic patients was effective in promoting significant regression of low-density non-calcified coronary atheroma regression as assessed by computed coronary angiography. This therapy has received Food and Drug Administration approval. Commonwealth Serum Laboratories has carried out a large clinical endpoint trial using an HDL complex (native apoA-I with phospholipid), and the results were negative. Our purpose is to review animal and human studies using various forms of HDL infusion therapy to promote regression of atherosclerosis. In our view, differences in results may be due to: 1) the HDL preparations used, 2) the subjects studied, and 3) the methods used to assess coronary atherosclerosis.


Sujet(s)
Lipoprotéines HDL , Humains , Animaux , Lipoprotéines HDL/administration et posologie , Lipoprotéines HDL/sang , Maladie des artères coronaires/traitement médicamenteux , Maladie des artères coronaires/imagerie diagnostique , Apolipoprotéine A-I/administration et posologie
3.
N Engl J Med ; 390(17): 1560-1571, 2024 May 02.
Article de Anglais | MEDLINE | ID: mdl-38587254

RÉSUMÉ

BACKGROUND: Cardiovascular events frequently recur after acute myocardial infarction, and low cholesterol efflux - a process mediated by apolipoprotein A1, which is the main protein in high-density lipoprotein - has been associated with an increased risk of cardiovascular events. CSL112 is human apolipoprotein A1 derived from plasma that increases cholesterol efflux capacity. Whether infusions of CSL112 can reduce the risk of recurrent cardiovascular events after acute myocardial infarction is unclear. METHODS: We conducted an international, double-blind, placebo-controlled trial involving patients with acute myocardial infarction, multivessel coronary artery disease, and additional cardiovascular risk factors. Patients were randomly assigned to receive either four weekly infusions of 6 g of CSL112 or matching placebo, with the first infusion administered within 5 days after the first medical contact for the acute myocardial infarction. The primary end point was a composite of myocardial infarction, stroke, or death from cardiovascular causes from randomization through 90 days of follow-up. RESULTS: A total of 18,219 patients were included in the trial (9112 in the CSL112 group and 9107 in the placebo group). There was no significant difference between the groups in the risk of a primary end-point event at 90 days of follow-up (439 patients [4.8%] in the CSL112 group vs. 472 patients [5.2%] in the placebo group; hazard ratio, 0.93; 95% confidence interval [CI], 0.81 to 1.05; P = 0.24), at 180 days of follow-up (622 patients [6.9%] vs. 683 patients [7.6%]; hazard ratio, 0.91; 95% CI, 0.81 to 1.01), or at 365 days of follow-up (885 patients [9.8%] vs. 944 patients [10.5%]; hazard ratio, 0.93; 95% CI, 0.85 to 1.02). The percentage of patients with adverse events was similar in the two groups; a higher number of hypersensitivity events was reported in the CSL112 group. CONCLUSIONS: Among patients with acute myocardial infarction, multivessel coronary artery disease, and additional cardiovascular risk factors, four weekly infusions of CSL112 did not result in a lower risk of myocardial infarction, stroke, or death from cardiovascular causes than placebo through 90 days. (Funded by CSL Behring; AEGIS-II ClinicalTrials.gov number, NCT03473223.).


Sujet(s)
Apolipoprotéine A-I , Lipoprotéines HDL , Infarctus du myocarde , Sujet âgé , Femelle , Humains , Mâle , Adulte d'âge moyen , Apolipoprotéine A-I/administration et posologie , Apolipoprotéine A-I/sang , Maladies cardiovasculaires/étiologie , Maladies cardiovasculaires/métabolisme , Maladies cardiovasculaires/mortalité , Maladies cardiovasculaires/prévention et contrôle , Maladie des artères coronaires/traitement médicamenteux , Maladie des artères coronaires/complications , Méthode en double aveugle , Perfusions veineuses , Estimation de Kaplan-Meier , Lipoprotéines HDL/sang , Lipoprotéines HDL/métabolisme , Infarctus du myocarde/complications , Infarctus du myocarde/traitement médicamenteux , Infarctus du myocarde/métabolisme , Infarctus du myocarde/mortalité , Récidive , Prévention secondaire , Accident vasculaire cérébral/prévention et contrôle , Facteurs de risque
4.
J Diabetes Res ; 2021: 6668506, 2021.
Article de Anglais | MEDLINE | ID: mdl-34095317

RÉSUMÉ

BACKGROUND AND AIMS: Apolipoprotein A-I (ApoA-I), the main component of high-density lipoprotein (HDL), not only promotes reverse cholesterol transport (RCT) in atherosclerosis but also increases insulin secretion in pancreatic ß-cells, suggesting that interventions which raise HDL levels may be beneficial in diabetes-associated cardiovascular disease (CVD). Previously, we showed that TNF-related apoptosis-inducing ligand (TRAIL) deletion in Apolipoprotein Eknockout (Apoe-/- ) mice results in diabetes-accelerated atherosclerosis in response to a "Western" diet. Here, we sought to identify whether reconstituted HDL (rHDL) could improve features of diabetes-associated CVD in Trail-/-Apoe-/- mice. METHODS AND RESULTS: Trail-/-Apoe-/- and Apoe-/- mice on a "Western" diet for 12 weeks received 3 weekly infusions of either PBS (vehicle) or rHDL (containing ApoA-I (20 mg/kg) and 1-palmitoyl-2-linoleoyl phosphatidylcholine). Administration of rHDL reduced total plasma cholesterol, triglyceride, and glucose levels in Trail-/-Apoe-/- but not in Apoe-/- mice, with no change in weight gain observed. rHDL treatment also improved glucose clearance in response to insulin and glucose tolerance tests. Immunohistological analysis of pancreata revealed increased insulin expression/production and a reduction in macrophage infiltration in mice with TRAIL deletion. Furthermore, atherosclerotic plaque size in Trail-/-Apoe-/- mice was significantly reduced associating with increased expression of the M2 macrophage marker CD206, suggesting HDL's involvement in the polarization of macrophages. rHDL also increased vascular mRNA expression of RCT transporters, ABCA1 and ABCG1, in Trail-/-Apoe-/- but not in Apoe-/- mice. Conclusions. rHDL improves features of diabetes-associated atherosclerosis in mice. These findings support the therapeutic potential of rHDL in the treatment of atherosclerosis and associated diabetic complications. More studies are warranted to understand rHDL's mechanism of action.


Sujet(s)
Anticholestérolémiants/administration et posologie , Athérosclérose/traitement médicamenteux , Glycémie/effets des médicaments et des substances chimiques , Cholestérol/sang , Diabète/traitement médicamenteux , Dyslipidémies/traitement médicamenteux , Hypoglycémiants/administration et posologie , Lipoprotéines HDL/administration et posologie , Membre-1 de la sous-famille A des transporteurs à cassette liant l'ATP/génétique , Membre-1 de la sous-famille A des transporteurs à cassette liant l'ATP/métabolisme , Membre-1 de la sous-famille G des transporteurs à cassette liant l'ATP/génétique , Membre-1 de la sous-famille G des transporteurs à cassette liant l'ATP/métabolisme , Animaux , Apolipoprotéine A-I/administration et posologie , Athérosclérose/sang , Athérosclérose/génétique , Marqueurs biologiques/sang , Glycémie/métabolisme , Diabète/sang , Régime occidental , Modèles animaux de maladie humaine , Dyslipidémies/sang , Dyslipidémies/génétique , Homéostasie , Humains , Macrophages/effets des médicaments et des substances chimiques , Macrophages/métabolisme , Mâle , Souris invalidées pour les gènes ApoE , Phosphatidylcholines/administration et posologie , Plaque d'athérosclérose , Ligand TRAIL/génétique , Ligand TRAIL/métabolisme
5.
Chem Res Toxicol ; 34(6): 1672-1680, 2021 06 21.
Article de Anglais | MEDLINE | ID: mdl-33861588

RÉSUMÉ

We previously demonstrated that apolipoprotein A-I (apoA-I), the major protein component of high-density lipoprotein (HDL), is an important target for myeloperoxidase (MPO)-catalyzed tyrosine chlorination in the circulation of subjects with cardiovascular diseases. Oxidation of apoA-I by MPO has been reported to deprive HDL of its protective properties. However, the potential effects of MPO-mediated site-specific tyrosine chlorination of apoA-I on dysfunctional HDL formation and atherosclerosis was unclear. Herein, Tyr192 in apoA-I was found to be the major chlorination site in both lesion and plasma HDL from humans with atherosclerosis, while MPO binding to apoA-I was demonstrated by immunoprecipitation studies in vivo. In vitro, MPO-mediated damage of lipid-free apoA-I impaired its ability to promote cellular cholesterol efflux by the ABCA1 pathway, whereas oxidation to lipid-associated apoA-I inhibited lecithin:cholesterol acyltransferase activation, two key steps in reverse cholesterol transport. Compared with native apoA-I, apoA-I containing a Tyr192 → Phe mutation was moderately resistant to oxidative inactivation by MPO. In high-fat-diet-fed apolipoprotein E-deficient mice, compared with native apoA-I, subcutaneous injection with oxidized apoA-I (MPO treated) failed to mediate the lipid content in aortic plaques while mutant apoA-I (Tyr192 → Phe) showed a slightly stronger ability to reduce the lipid content in vivo. Our observations suggest that oxidative damage of apoA-I and HDL involves MPO-dependent site-specific tyrosine chlorination, raising the feasibility of producing MPO-resistant forms of apoA-I that have stronger antiatherosclerotic activity in vivo.


Sujet(s)
Apolipoprotéine A-I/métabolisme , Lipoprotéines HDL/métabolisme , Myeloperoxidase/métabolisme , Adulte , Animaux , Apolipoprotéine A-I/administration et posologie , Apolipoprotéine A-I/sang , Athérosclérose , Halogénation , Humains , Injections sous-cutanées , Souris , Souris invalidées pour les gènes ApoE , Myeloperoxidase/sang , Tyrosine/métabolisme
6.
Circ Res ; 127(11): 1422-1436, 2020 11 06.
Article de Anglais | MEDLINE | ID: mdl-32951519

RÉSUMÉ

RATIONALE: Decades of research have examined immune-modulatory strategies to protect the heart after an acute myocardial infarction and prevent progression to heart failure but have failed to translate to clinical benefit. OBJECTIVE: To determine anti-inflammatory actions of n-apo AI (Apo AI nanoparticles) that contribute to cardiac tissue recovery after myocardial infarction. METHODS AND RESULTS: Using a preclinical mouse model of myocardial infarction, we demonstrate that a single intravenous bolus of n-apo AI (CSL111, 80 mg/kg) delivered immediately after reperfusion reduced the systemic and cardiac inflammatory response. N-apo AI treatment lowered the number of circulating leukocytes by 30±7% and their recruitment into the ischemic heart by 25±10% (all P<5.0×10-2). This was associated with a reduction in plasma levels of the clinical biomarker of cardiac injury, cardiac troponin-I, by 52±17% (P=1.01×10-2). N-apo AI reduced the cardiac expression of chemokines that attract neutrophils and monocytes by 60% to 80% and lowered surface expression of integrin CD11b on monocytes by 20±5% (all P<5.0×10-2). Fluorescently labeled n-apo AI entered the infarct and peri-infarct regions and colocalized with cardiomyocytes undergoing apoptosis and with leukocytes. We further demonstrate that n-apo AI binds to neutrophils and monocytes, with preferential binding to the proinflammatory monocyte subtype and partially via SR-BI (scavenger receptor BI). In patients with type 2 diabetes, we also observed that intravenous infusion of the same n-apo AI (CSL111, 80 mg/kg) similarly reduced the level of circulating leukocytes by 12±5% (all P<5.0×10-2). CONCLUSIONS: A single intravenous bolus of n-apo AI delivered immediately post-myocardial infarction reduced the systemic and cardiac inflammatory response through direct actions on both the ischemic myocardium and leukocytes. These data highlight the anti-inflammatory effects of n-apo AI and provide preclinical support for investigation of its use for management of acute coronary syndromes in the setting of primary percutaneous coronary interventions.


Sujet(s)
Anti-inflammatoires/administration et posologie , Apolipoprotéine A-I/administration et posologie , Inflammation/prévention et contrôle , Leucocytes/effets des médicaments et des substances chimiques , Infarctus du myocarde/traitement médicamenteux , Nanoparticules , Administration par voie intraveineuse , Adulte , Animaux , Antigènes CD11b/métabolisme , Cellules cultivées , Chimiokines/métabolisme , Diabète de type 2/sang , Diabète de type 2/traitement médicamenteux , Diabète de type 2/immunologie , Modèles animaux de maladie humaine , Calendrier d'administration des médicaments , Humains , Inflammation/immunologie , Inflammation/métabolisme , Leucocytes/immunologie , Leucocytes/métabolisme , Mâle , Souris de souche-129 , Souris de lignée C57BL , Souris knockout , Adulte d'âge moyen , Infarctus du myocarde/immunologie , Infarctus du myocarde/métabolisme , Infarctus du myocarde/anatomopathologie , Essais contrôlés randomisés comme sujet , Récepteurs éboueurs de classe B/génétique , Récepteurs éboueurs de classe B/métabolisme , Troponine I/sang
7.
Commun Biol ; 3(1): 386, 2020 07 16.
Article de Anglais | MEDLINE | ID: mdl-32678293

RÉSUMÉ

Many patients of choroidal neovascularization (CNV) are unresponsive to the current anti-VEGF treatment. The mechanisms for anti-VEGF resistance are poorly understood. We explore the unique property of the apolipoprotein A-I (apoA-I) binding protein (AIBP) that enhances cholesterol efflux from endothelial cells and macrophages to thereby limit angiogenesis and inflammation to tackle anti-VEGF resistance in CNV. We show that laser-induced CNV in mice with increased age showed increased resistance to anti-VEGF treatment, which correlates with increased lipid accumulation in macrophages. The combination of AIBP/apoA-I and anti-VEGF treatment overcomes anti-VEGF resistance and effectively suppresses CNV. Furthermore, macrophage depletion in old mice restores CNV sensitivity to anti-VEGF treatment and blunts the synergistic effect of combination therapy. These results suggest that cholesterol-laden macrophages play a critical role in inducing anti-VEGF resistance in CNV. Combination therapy by neutralizing VEGF and enhancing cholesterol removal from macrophages is a promising strategy to combat anti-VEGF resistance in CNV.


Sujet(s)
Apolipoprotéine A-I/usage thérapeutique , Néovascularisation choroïdienne/traitement médicamenteux , Phosphoprotéines/usage thérapeutique , Racémases et épimérases/usage thérapeutique , Facteur de croissance endothéliale vasculaire de type A/antagonistes et inhibiteurs , Sujet âgé , Sujet âgé de 80 ans ou plus , Animaux , Apolipoprotéine A-I/administration et posologie , Membrane cellulaire/métabolisme , Cholestérol/métabolisme , Choroïde/métabolisme , Résistance aux substances , Association de médicaments , Femelle , Humains , Macrophages/effets des médicaments et des substances chimiques , Mâle , Souris , Néovascularisation pathologique/traitement médicamenteux , Phosphoprotéines/administration et posologie , Racémases et épimérases/administration et posologie , Rétine/métabolisme
8.
Mol Metab ; 35: 100949, 2020 05.
Article de Anglais | MEDLINE | ID: mdl-32244181

RÉSUMÉ

OBJECTIVE: Acute administration of the main protein component of high-density lipoprotein, apolipoprotein A-I (ApoA-1), improves glucose uptake in skeletal muscle. The molecular mechanisms mediating this are not known, but in muscle cell cultures, ApoA-1 failed to increase glucose uptake when infected with a dominant-negative AMP-activated protein kinase (AMPK) virus. We therefore investigated whether AMPK is necessary for ApoA-1-stimulated glucose uptake in intact heart and skeletal muscle in vivo. METHODS: The effect of injection with recombinant human ApoA-1 (rApoA-1) on glucose tolerance, glucose-stimulated insulin secretion, and glucose uptake into skeletal and heart muscle with and without block of insulin secretion by injection of epinephrine (0.1 mg/kg) and propranolol (5 mg/kg), were investigated in 8 weeks high-fat diet-fed (60E%) wild-type and AMPKα2 kinase-dead mice in the overnight-fasted state. In addition, the effect of rApoA-1 on glucose uptake in isolated skeletal muscle ex vivo was studied. RESULTS: rApoA-1 lowered plasma glucose concentration by 1.7 mmol/l within 3 h (6.1 vs 4.4 mmol/l; p < 0.001). Three hours after rApoA-1 injection, glucose tolerance during a 40-min glucose tolerance test (GTT) was improved compared to control (area under the curve (AUC) reduced by 45%, p < 0.001). This was accompanied by an increased glucose clearance into skeletal (+110%; p < 0.001) and heart muscle (+100%; p < 0.001) and an increase in glucose-stimulated insulin secretion 20 min after glucose injection (+180%; p < 0.001). When insulin secretion was blocked during a GTT, rApoA-1 still enhanced glucose tolerance (AUC lowered by 20% compared to control; p < 0.001) and increased glucose clearance into skeletal (+50%; p < 0.05) and heart muscle (+270%; p < 0.001). These improvements occurred to a similar extent in both wild-type and AMPKα2 kinase-dead mice and thus independently of AMPKα2 activity in skeletal- and heart muscle. Interestingly, rApoA-1 failed to increase glucose uptake in isolated skeletal muscles ex vivo. CONCLUSIONS: In conclusion, ApoA-1 stimulates in vivo glucose disposal into skeletal and heart muscle independently of AMPKα2. The observation that ApoA-1 fails to increase glucose uptake in isolated muscle ex vivo suggests that additional systemic effects are required.


Sujet(s)
AMP-Activated Protein Kinases/métabolisme , Apolipoprotéine A-I/administration et posologie , Glycémie/métabolisme , Muscles squelettiques/métabolisme , Myocarde/métabolisme , Transduction du signal/effets des médicaments et des substances chimiques , AMP-Activated Protein Kinases/génétique , Animaux , Alimentation riche en graisse , Femelle , Hyperglycémie provoquée , Insuline/métabolisme , Sécrétion d'insuline/effets des médicaments et des substances chimiques , Souris , Souris de lignée C57BL , Souris transgéniques , Protéines recombinantes/administration et posologie
9.
Drug Deliv ; 27(1): 530-541, 2020 Dec.
Article de Anglais | MEDLINE | ID: mdl-32241173

RÉSUMÉ

Reconstituted high density lipoprotein (rHDL) has been regarded as a promising brain-targeting vehicle for anti-glioma drugs under the mediation of apolipoprotein A-I (apoA-I). However, some stability issues relating to drug leakage and consequent reduced targeting efficiency in the course of discoidal rHDL (d-rHDL) circulating in blood hinder its broad application. The objective of the study was to develop a novel stabilized d-rHDL by replacing cholesterol and apoA-I with mono-cholesterol glutarate (MCG) modified apoA-I (termed as mA) and to evaluate its allosteric behavior and glioma targeting. MCG was synthesized through esterifying the hydroxyl of cholesterol with glutaric anhydride and characterized by FI-IR and 1H NMR. d-rHDL assembled with mA (termed as m-d-rHDL) presented similar properties such as minute particle size and disk-like appearance resembling nascent HDL. Morphological transformation observation and in vitro release plots convinced that the modification of cholesterol could effectively inhibit the remolding of d-rHDL. The uptake of m-d-rHDL by LCAT-pretreated bEND.3 cells was significantly higher than that of d-rHDL, thereby serving as another proof for the capability of m-d-rHDL in enhancing targeting property. Besides, apoA-I anchoring into m-d-rHDL played a critical role in the endocytosis process into bEND.3 cells and C6 cells, which implied the possibility of traversing blood brain barrier and accumulating in the brain and glioma. These results suggested that the modification toward cholesterol to improve the stability of d-rHDL is advantageous, and that this obtained m-d-rHDL revealed great potential for realization of suppressing the remolding of d-rHDL in the brain-targeted treatment of glioma for drug delivery.


Sujet(s)
Apolipoprotéine A-I/composition chimique , Apolipoprotéine A-I/métabolisme , Gliome/traitement médicamenteux , Lipoprotéines HDL/composition chimique , Lipoprotéines HDL/métabolisme , Animaux , Apolipoprotéine A-I/administration et posologie , Transport biologique , Lignée cellulaire , Lignée cellulaire tumorale , Vecteurs de médicaments/administration et posologie , Vecteurs de médicaments/composition chimique , Vecteurs de médicaments/métabolisme , Systèmes de délivrance de médicaments , Gliome/métabolisme , Glutarates/administration et posologie , Glutarates/métabolisme , Humains , Lipoprotéines HDL/administration et posologie , Lipoprotéines HDL/synthèse chimique , Souris , Taille de particule , Rats
10.
J Pharmacol Exp Ther ; 372(1): 54-62, 2020 01.
Article de Anglais | MEDLINE | ID: mdl-31649050

RÉSUMÉ

Obesity is a pathologic condition generated by an energy imbalance, that is, excess caloric consumption, leading to weight gain and metabolic disturbances characterized by adipose tissue inflammation and hyperglycemic conditions. In line with these observations, increasing evidence causally links inflammation, or the molecules and networks integral to inflammatory response, to the development of obesity and the complications that emerge from this pathology, such as cardiovascular, neurologic, respiratory, and metabolic illnesses, as well as sepsis and cancer. Not surprisingly, this chronic and abnormal metabolic background leads to constant derangements in innate and adaptive immunity. It is well known that high-density lipoprotein (HDL) possesses anti-inflammatory and antioxidant properties, and various studies have highlighted an emerging role of HDL in modulating immune function. The efficacy of synthetic HDL (sHDL) containing the recombinant form of apoA-IMilano (sHDL-apoA-IM), originating from the observation that carriers of this mutation have low levels of HDL cholesterol without increased atherosclerosis, has been largely proved in diverse animal models of atherosclerosis; however, the therapeutic use of sHDL-apoA-IM still needs clinical validation. One of the main limitations to the use of recombinant proteins in clinical studies lies in the unsustainable purification costs. Unpurified rice-milk-apoA-IM demonstrated anti-inflammatory and antiatherogenic properties in a mouse model, even though administrated by an unconventional way: by oral gavage. Additionally, recent data have uncovered new therapeutic applications for this sHDL-apoA-IM This review provides an overview of all potential application of sHDL-apoA-IM in some inflammatory-based diseases. SIGNIFICANCE STATEMENT: A recent study demonstrated that oral administration of rice-seed protein extracts containing the apoA-IM (i.e., the milk-apoA-IM) reduced atherosclerosis development in a mouse model. Moreover, the rice-milk-apoA-IM preserved both in vitro and in vivo anti-inflammatory properties, as observed when sHDL-apoA-IM was given by intravascular infusion. Besides, various studies suggested that sHDL-apoA-IM could positively affect other inflammatory-based diseases. Together, these data might represent a new starting point for "sHDL-apoA-IM-based therapies" in chronic degenerative disease.


Sujet(s)
Anti-inflammatoires/usage thérapeutique , Anticholestérolémiants/usage thérapeutique , Apolipoprotéine A-I/usage thérapeutique , Athérosclérose/traitement médicamenteux , Animaux , Anti-inflammatoires/administration et posologie , Anticholestérolémiants/administration et posologie , Apolipoprotéine A-I/administration et posologie , Athérosclérose/prévention et contrôle , Humains
11.
Can J Cardiol ; 35(10): 1400-1408, 2019 10.
Article de Anglais | MEDLINE | ID: mdl-31495683

RÉSUMÉ

BACKGROUND: Among strategies to reduce the remaining risk of cardiovascular disease, interest has focused on using infusions of synthetic high-density lipoprotein (sHDL). METHODS: New Zealand rabbits underwent a perivascular injury at both carotids and were randomly allocated into 2 protocols: (1) a single-dose study, where rabbits were treated with a single infusion of sHDL containing a trimeric form of human apoA-I (TN-sHDL, 200 mg/kg) or with Placebo; (2) a multiple-dose study, where 4 groups of rabbits were treated 5 times with Placebo or TN-sHDL at different doses (8, 40, 100 mg/kg). Plaque changes were analysed in vivo by intravascular ultrasound. Blood was drawn from rabbits for biochemical analyses and cholesterol efflux capacity evaluation. RESULTS: In both protocols, atheroma volume in the Placebo groups increased between the first and the second intravascular ultrasound evaluation. A stabilization or a slight regression was instead observed vs baseline in the TN-sHDL-treated groups (P < 0.005 vs Placebo after infusion). TN-sHDL treatment caused a sharp rise of plasma-free cholesterol levels and a significant increase of total cholesterol efflux capacity. Histologic analysis of carotid plaques showed a reduced macrophage accumulation in TN-sHDL-treated rabbits compared with Placebo (P < 0.05). CONCLUSIONS: Our results demonstrate that acute and subacute treatments with TN-sHDL are effective in stabilizing atherosclerotic plaques in a rabbit model. This effect appears to be related to a reduced intraplaque accumulation of inflammatory cells. Besides recent failures in proving its efficacy, sHDL treatment remains a fascinating therapeutic option for the reduction of cardiovascular risk.


Sujet(s)
Apolipoprotéine A-I/administration et posologie , Lipoprotéines HDL/administration et posologie , Plaque d'athérosclérose/prévention et contrôle , Animaux , Hypercholestérolémie/complications , Perfusions veineuses , Mâle , Préparations pharmaceutiques , Plaque d'athérosclérose/étiologie , Lapins , Répartition aléatoire
12.
Arterioscler Thromb Vasc Biol ; 39(6): 1160-1171, 2019 06.
Article de Anglais | MEDLINE | ID: mdl-31018664

RÉSUMÉ

Objective- Insulin resistance and inflammation in pregnancy are risk factors for gestational diabetes mellitus. Increased plasma HDL (high-density lipoprotein) and apo (apolipoprotein) A-I levels have been reported to improve glucose metabolism and inhibit inflammation in animals and humans. This study asks whether increasing plasma apoA-I levels improves insulin sensitivity and reduces inflammation in insulin-resistant pregnant rats. Approach and Results- Insulin-resistant pregnant rats received intravenous infusions of lipid-free apoA-I (8 mg/kg) or saline on days 6, 9, 12, 15, and 18 of pregnancy. The rats were then subjected to a euglycemic-hyperinsulinemic clamp. Glucose uptake was increased in white and brown adipose tissue by 57±13% and 32±10%, respectively ( P<0.05 for both), and in quadriceps and gastrocnemius muscle by 35±9.7% and 47±14%, respectively ( P<0.05 for both), in the apoA-I-treated pregnant rats relative to saline-infused pregnant rats. The pregnant rats that were treated with apoA-I also had reduced plasma TNF-α (tumor necrosis factor-α) levels by 57±8.4%, plasma IL (interleukin)-6 levels by 67±9.5%, and adipose tissue macrophage content by 54±8.2% ( P<0.05 for all) relative to the saline-treated pregnant rats. Conclusions- These studies establish that apoA-I protects against pregnancy-induced insulin resistance in rats by increasing insulin sensitivity in adipose tissue and skeletal muscle and inhibiting inflammation. This identifies apoA-I as a potential target for preventing pregnancy-induced insulin resistance and reducing the incidence of gestational diabetes mellitus.


Sujet(s)
Anti-inflammatoires/administration et posologie , Apolipoprotéine A-I/administration et posologie , Glycémie/effets des médicaments et des substances chimiques , Diabète gestationnel/prévention et contrôle , Hypoglycémiants/administration et posologie , Insulinorésistance , Insuline/sang , Tissu adipeux/effets des médicaments et des substances chimiques , Tissu adipeux/métabolisme , Animaux , Marqueurs biologiques/sang , Glycémie/métabolisme , Diabète gestationnel/sang , Modèles animaux de maladie humaine , Femelle , Médiateurs de l'inflammation/sang , Perfusions veineuses , Interleukine-6/sang , Macrophages/effets des médicaments et des substances chimiques , Macrophages/métabolisme , Grossesse , Muscle quadriceps fémoral/effets des médicaments et des substances chimiques , Muscle quadriceps fémoral/métabolisme , Rat Wistar , Facteur de nécrose tumorale alpha/sang
13.
J Control Release ; 296: 14-28, 2019 02 28.
Article de Anglais | MEDLINE | ID: mdl-30639387

RÉSUMÉ

Alzheimer's disease (AD) is a multifaceted and progressive neurodegenerative disease characterized by accumulation of amyloid-beta (Aß) and deficits of acetylcholine. Accordingly, the intra-/extra-cerebral level of high density lipoprotein (HDL) is crucial on the pathogenesis of AD; and most of all, various HDL-protein subtypes play a double-edged role in AD pathology, of which apolipoprotein A-I (apoA-I) gives protective outcomes. Inspired from "HDL bionics", we proposed biologically reassembled nanodrugs, donepezil-loaded apolipoprotein A-I-reconstituted HDL (rHDL/Do) that concurrently executed dual-missions of Aß-targeting clearance and acetylcholinesterase (AChE) inhibition in AD therapy. Once prepared, rHDL/Do nanodrug achieved high drug encapsulation efficiency of 90.47%, and mimicked the configurations and properties of natural lipoproteins aiming to significantly enhance BBB penetration and modulate Aß-induced neuronal damage both in vitro and in vivo. Surface plasmon resonance (SPR) analysis confirmed that rHDL/Do facilitated microglial-mediated Aß intake and degradation, demonstrating low KD value with Aß affinity (2.45 × 10-8 of Aß monomer and 2.78 × 10-8 of Aß oligomer). In AD animal models, daily treatment of rHDL/Do efficiently inhibited AChE activity, ameliorated neurologic variation, promoted Aß clearance, and rescued memory loss at a safe level. The collective findings indicated that the biological nanodrug was provided with the capacities of BBB penetration, Aß capture and degradation via microglial cells, and cholinergic dysfunction amelioration after controlled donepezil release. In summary, rHDL/Do nanodrugs could offer a promising strategy to synergize both symptom control and disease modification in AD therapy.


Sujet(s)
Maladie d'Alzheimer/traitement médicamenteux , Apolipoprotéine A-I/administration et posologie , Anticholinestérasiques/administration et posologie , Donépézil/administration et posologie , Lipoprotéines HDL/administration et posologie , Nanoparticules/administration et posologie , Neuroprotecteurs/administration et posologie , Maladie d'Alzheimer/métabolisme , Peptides bêta-amyloïdes/métabolisme , Animaux , Apolipoprotéine A-I/composition chimique , Barrière hémato-encéphalique/métabolisme , Lignée cellulaire , Anticholinestérasiques/composition chimique , Modèles animaux de maladie humaine , Donépézil/composition chimique , Libération de médicament , Humains , Lipoprotéines HDL/composition chimique , Lipoprotéines HDL/pharmacocinétique , Mâle , Souris de lignée ICR , Nanoparticules/composition chimique , Neuroprotecteurs/composition chimique , Fragments peptidiques/métabolisme , Rat Sprague-Dawley
14.
Br J Pharmacol ; 175(21): 4167-4182, 2018 11.
Article de Anglais | MEDLINE | ID: mdl-30079544

RÉSUMÉ

BACKGROUND AND PURPOSE: The pleiotropic properties of HDL may exert beneficial effects on the myocardium. The effect of recombinant HDLMilano on established heart failure was evaluated in C57BL/6 mice. EXPERIMENTAL APPROACH: Mice were subjected to transverse aortic constriction (TAC) or sham operation at the age of 14 weeks. Eight weeks later, TAC and sham mice were each randomized into three different groups. Reference groups were killed at day 56 after the operation for baseline analysis. Five i.p. injections of recombinant HDLMilano (MDCO-216), 100 mg·kg-1 , or an equivalent volume of control buffer were administered with a 48 h interval starting at day 56. Endpoint analyses in the control buffer groups and in the MDCO-216 groups were executed at day 65. KEY RESULTS: Lung weight in MDCO-216 TAC mice was 25.3% lower than in reference TAC mice and 27.9% lower than in control buffer TAC mice and was similar in MDCO-216 sham mice. MDCO-216 significantly decreased interstitial fibrosis and increased relative vascularity compared to reference TAC mice and control buffer TAC mice. The peak rate of isovolumetric relaxation in MDCO-216 TAC mice was 30.4 and 36.3% higher than in reference TAC mice and control buffer TAC mice respectively. Nitro-oxidative stress and myocardial apoptosis were significantly reduced in MDCO-216 TAC mice compared to control buffer TAC mice. CONCLUSIONS AND IMPLICATIONS: MDCO-216 improves diastolic function, induces regression of interstitial fibrosis and normalizes lung weight in mice with established heart failure. Recombinant HDL may emerge as a treatment modality in heart failure.


Sujet(s)
Apolipoprotéine A-I/usage thérapeutique , Défaillance cardiaque/traitement médicamenteux , Phosphatidylcholines/usage thérapeutique , Animaux , Apolipoprotéine A-I/administration et posologie , Association médicamenteuse , Défaillance cardiaque/métabolisme , Défaillance cardiaque/anatomopathologie , Injections péritoneales , Lipides/sang , Souris , Souris de lignée C57BL , Phosphatidylcholines/administration et posologie
15.
JAMA Cardiol ; 3(9): 815-822, 2018 09 01.
Article de Anglais | MEDLINE | ID: mdl-30046828

RÉSUMÉ

Importance: CER-001 is a negatively charged, engineered pre-ß high-density lipoprotein (HDL) mimetic containing apolipoprotein A-I and sphingomyelin. Preliminary studies demonstrated favorable effects of CER-001 on cholesterol efflux and vascular inflammation. A post hoc reanalysis of a previously completed study of intravenous infusion of CER-001, 3 mg/k, showed that the intravenous infusion in patients with a high coronary plaque burden promoted regression as assessed by intravascular ultrasonography. Objective: To determine the effect of infusing CER-001 on coronary atherosclerosis progression in statin-treated patients. Design, Setting, and Participants: A double-blind, randomized, multicenter trial evaluating the effect of 10 weekly intravenous infusions of CER-001, 3 mg/kg, (n = 135) or placebo (n = 137) in patients with an acute coronary syndrome (ACS) and baseline percent atheroma volume (PAV) greater than 30% in the proximal segment of an epicardial artery by intravascular ultrasonography. The study included 34 academic and community hospitals in Australia, Hungary, the Netherlands, and the United States in patients with ACS presenting for coronary angiography. Patients were enrolled from August 15, 2015, to November 19, 2016. Interventions: Participants were randomized to receive weekly CER-001, 3 mg/kg, or placebo for 10 weeks in addition to statins. Main Outcomes and Measures: The primary efficacy measure was the nominal change in PAV from baseline to day 78 measured by serial intravascular ultrasonography imaging. The secondary efficacy measures were nominal change in normalized total atheroma volume and percentage of patients demonstrating plaque regression. Safety and tolerability were also evaluated. Results: Among 293 patients (mean [SD] age, 59.8 [9.4] years; 217 men [79.8%] and 261 white race/ethnicity [96.0%]), 86 (29%) had statin prior use prior to the index ACS and 272 (92.8%) had evaluable imaging at follow-up. The placebo and CER-001 groups had similar posttreatment median levels of low-density lipoprotein cholesterol (74 mg/dL vs 79 mg/dL; P = .15) and high-density lipoprotein cholesterol (43 mg/dL vs 44 mg/dL; P = .66). The primary efficacy measure, PAV, decreased 0.41% with placebo (P = .005 compared with baseline), but not with CER-001 (-0.09%; P = .67 compared with baseline; between group differences, 0.32%; P = .15). Similar percentages of patients in the placebo and CER-001 groups demonstrated regression of PAV (57.7% vs 53.3%; P = .49). Infusions were well tolerated, with no differences in clinical and laboratory adverse events observed between treatment groups. Conclusions and Relevance: Infusion of CER-001 did not promote regression of coronary atherosclerosis in statin-treated patients with ACS and high plaque burden. Trial Registration: ClinicalTrials.gov Identifier: NCT2484378.


Sujet(s)
Syndrome coronarien aigu/imagerie diagnostique , Syndrome coronarien aigu/traitement médicamenteux , Apolipoprotéine A-I/administration et posologie , Maladie des artères coronaires/imagerie diagnostique , Maladie des artères coronaires/traitement médicamenteux , Inhibiteurs de l'hydroxyméthylglutaryl-CoA réductase/administration et posologie , Phospholipides/administration et posologie , Protéines recombinantes/administration et posologie , Sujet âgé , Apolipoprotéine A-I/usage thérapeutique , Australie , Évolution de la maladie , Méthode en double aveugle , Calendrier d'administration des médicaments , Femelle , Humains , Hongrie , Inhibiteurs de l'hydroxyméthylglutaryl-CoA réductase/usage thérapeutique , Pompes à perfusion , Mâle , Adulte d'âge moyen , Pays-Bas , Phospholipides/usage thérapeutique , Protéines recombinantes/usage thérapeutique , Résultat thérapeutique , Échographie interventionnelle , États-Unis
16.
JAMA Cardiol ; 3(9): 806-814, 2018 09 01.
Article de Anglais | MEDLINE | ID: mdl-30046837

RÉSUMÉ

Importance: Infusing a high-density lipoprotein mimetic containing apolipoprotein A-I Milano demonstrated potential atheroma regression in patients following an acute coronary syndrome. To our knowledge, the effect of infusing a new mimetic preparation (MDCO-216) with contemporary statin therapy is unknown. Objective: To determine the effect of infusing MDCO-216 on coronary atherosclerosis progression. Design, Setting, and Participants: This double-blind, randomized clinical trial conducted in 22 hospitals in Canada and Europe compared the effects of 5 weekly intravenous infusions of MDCO-216 at a dose of 20 mg/kg weekly (n = 59) with placebo (n = 67) in statin-treated patients with an acute coronary syndrome. Main Outcomes and Measures: The primary efficacy measure was the nominal change in percent atheroma volume (PAV) from baseline to day 36 as measured by serial intravascular ultrasonography. The secondary efficacy measures were the nominal changes in normalized total atheroma volume (TAV), atheroma volume in the most diseased 10-mm segment, and the percentage of patients who demonstrated plaque regression. Safety and tolerability were also evaluated. Results: Among 122 randomized patients (mean [SD] age, 61.8 [10.4] years; 93 men [76.2%]; 61 [50.0%] with prior statin use; and a mean [SD] low-density lipoprotein cholesterol [LDL-C] level of 87.6 [40.5] mg/dL [to convert to millimoles per liter, multiply by 0.0259]), 113 (92.6%) had evaluable imaging results at follow-up. The receiving-treatment LDL-C levels were comparable with the placebo and MDCO-216 (68.6 vs 70.5 mg/dL; difference, -2.5 mg/dL; 95% CI, -10.1 to 5.0; P = .51). A reduction in high-density lipoprotein cholesterol levels was observed in MDCO, but not placebo patients (-3.3 vs 3.0 mg/dL [to convert to millimoles per liter, multiply by 0.0259]; difference, -6.3 mg/dL; 95% CI, -8.5 to -4.1; P < .001). Percent atheroma volume, which was adjusted for baseline values, decreased 0.94% with the placebo and 0.21% with MDCO-216 (difference, 0.73%; 95% CI, -0.07 to 1.52; P = .07). Normalized TAV decreased 7.9 mm3 with the placebo and 6.4 mm3 with MDCO-216 (difference, 1.6 mm3; 95% CI, -5.6 to 8.7; P = .67), and atheroma volume in the most diseased segment decreased 1.8 mm3 with the placebo and 2.2 mm3 with MDCO-216 (difference 0.4 mm3; 95% CI, -4.4 to 3.5; P = .83). A similar percentage of patients demonstrated a regression of PAV (67.2% vs 55.8%; P = .21) and TAV (68.9% vs 71.2%; P = .79) in the placebo and MDCO-216 groups, respectively. Conclusions and Relevance: Among patients with an acute coronary syndrome, infusing MDCO-216 did not produce an incremental plaque regression in the setting of contemporary statin therapy. Trial Registration: ClinicalTrials.gov Identifier: NCT02678923.


Sujet(s)
Syndrome coronarien aigu/traitement médicamenteux , Apolipoprotéine A-I/administration et posologie , Cholestérol LDL/métabolisme , Maladie des artères coronaires/traitement médicamenteux , Inhibiteurs de l'hydroxyméthylglutaryl-CoA réductase/administration et posologie , Syndrome coronarien aigu/imagerie diagnostique , Syndrome coronarien aigu/métabolisme , Administration par voie intraveineuse , Sujet âgé , Apolipoprotéine A-I/usage thérapeutique , Canada , Maladie des artères coronaires/imagerie diagnostique , Maladie des artères coronaires/métabolisme , Méthode en double aveugle , Calendrier d'administration des médicaments , Europe , Femelle , Humains , Inhibiteurs de l'hydroxyméthylglutaryl-CoA réductase/usage thérapeutique , Pompes à perfusion , Mâle , Adulte d'âge moyen , Projets pilotes , Résultat thérapeutique , Échographie interventionnelle
17.
Int J Cardiol ; 271: 233-239, 2018 Nov 15.
Article de Anglais | MEDLINE | ID: mdl-29907443

RÉSUMÉ

BACKGROUND: Atherosclerosis is a slowly progressing, chronic multifactorial disease characterized by the accumulation of lipids, inflammatory cells, and fibrous tissue that drives to the formation of asymmetric focal thickenings in the tunica intima of large and mid-sized arteries. Despite the high therapeutic potential of ApoA-1 proteins, the purification and delivery into the disordered organisms of these drugs is still limited by low efficiency in these processes. METHODS AND RESULTS: We report here a novel production and delivery system of anti-atherogenic APOA-1Milano muteins (APOA-1M) by means of genetically modified rice plants. APOA-1M, delivered as protein extracts from transgenic rice seeds, significantly reduced macrophage activation and foam cell formation in vitro in oxLDL-loaded THP-1 model. The APOA-1M delivery method and therapeutic efficacy was tested in healthy mice and in Apoe-/- mice fed with high cholesterol diet (Western Diet, WD). APOA-1M rice milk significantly reduced atherosclerotic plaque size and lipids composition in aortic sinus and aortic arch of WD-fed Apoe-/- mice as compared to wild type rice milk-treated, WD-fed Apoe-/- mice. APOA-1M rice milk also significantly reduced macrophage number in liver of WD-fed Apoe-/- mice as compared to WT rice milk treated mice. TRANSLATIONAL IMPACT: The delivery of therapeutic APOA-1M full length proteins via oral administration of rice seeds protein extracts (the 'rice milk') to the disordered organism, without any need of purification, might overcome the main APOA1-based therapies' limitations and improve the use of this molecules as therapeutic agents for cardiovascular patients.


Sujet(s)
Anti-inflammatoires/administration et posologie , Apolipoprotéine A-I/administration et posologie , Apolipoprotéines E/déficit , Athérosclérose/traitement médicamenteux , Oryza/génétique , Plaque d'athérosclérose/traitement médicamenteux , Administration par voie orale , Animaux , Athérosclérose/métabolisme , Athérosclérose/anatomopathologie , Relation dose-effet des médicaments , Aliment génétiquement modifié , Mâle , Souris , Souris knockout , Plaque d'athérosclérose/métabolisme , Plaque d'athérosclérose/anatomopathologie
18.
Ther Deliv ; 9(4): 257-268, 2018 03 01.
Article de Anglais | MEDLINE | ID: mdl-29495929

RÉSUMÉ

The physiological role(s) of mammalian plasma lipoproteins is to transport hydrophobic molecules (primarily cholesterol and triacylglycerols) to their respective destinations. Lipoproteins have also been studied as drug-delivery agents due to their advantageous payload capacity, long residence time in the circulation and biocompatibility. The purpose of this review is to briefly discuss current findings with the focus on each type of formulation's potential for clinical applications. Regarding utilizing lipoprotein type formulation for cancer therapeutics, their potential for tumor-selective delivery is also discussed.


Sujet(s)
Préparation de médicament/méthodes , Systèmes de délivrance de médicaments/méthodes , Lipoprotéines/composition chimique , Nanoparticules/composition chimique , Nanomédecine théranostique/méthodes , Animaux , Antinéoplasiques/administration et posologie , Apolipoprotéine A-I/administration et posologie , Athérosclérose/traitement médicamenteux , Matériaux biomimétiques/composition chimique , Essais cliniques comme sujet , Préparation de médicament/tendances , Systèmes de délivrance de médicaments/tendances , Humains , Tumeurs/traitement médicamenteux , Phospholipides/administration et posologie , Protéines recombinantes/administration et posologie , Nanomédecine théranostique/tendances
19.
Neurobiol Aging ; 60: 116-128, 2017 12.
Article de Anglais | MEDLINE | ID: mdl-28941727

RÉSUMÉ

Beyond the crucial role of apolipoprotein A-I (ApoA-I) on peripheral cholesterol metabolism, this apolipoprotein has also been implicated in beta amyloid (Aß)-related neuropathologies. ApoA-I-Milano (M) is a mutated variant, which showed increased vasoprotective properties compared to ApoA-I-wild type in models of atherosclerosis and cardiovascular damage. We speculated that ApoA-I-M may also protect Aß-affected vasculature and reverse some of the pathological features associated with Alzheimer's disease (AD). For this purpose, we produced and characterized human recombinant ApoA-I-wild type and ApoA-I-M proteins. Both of them were able to avoid the aggregation of Aß in vitro, even though recombinant ApoA-I-M was significantly more effective in protecting endothelial cells from Aß(1-42)-toxicity. Next, we determined the effect of chronic intravenous administration of rApoA-I-M in the APP23-transgenic mouse model of AD. We found reduced cerebral Aß levels in mice that received rApoA-I-M, which were accompanied by a lower expression of astrocyte and microglia neuroinflammatory markers. Our results suggest an applicability of this molecule as a therapeutic candidate for protecting the brain in AD.


Sujet(s)
Maladie d'Alzheimer/traitement médicamenteux , Maladie d'Alzheimer/métabolisme , Peptides bêta-amyloïdes/métabolisme , Apolipoprotéine A-I/administration et posologie , Encéphale/métabolisme , Animaux , Apolipoprotéine A-I/pharmacologie , Apolipoprotéine A-I/physiologie , Modèles animaux de maladie humaine , Perfusions veineuses , Souris transgéniques , Protéines recombinantes/administration et posologie , Protéines recombinantes/pharmacologie
20.
J Am Heart Assoc ; 6(9)2017 Sep 22.
Article de Anglais | MEDLINE | ID: mdl-28939717

RÉSUMÉ

BACKGROUND: Subcutaneously injected lipid-free apoA-I (apolipoprotein A-I) reduces accumulation of lipid and immune cells within the aortic root of hypercholesterolemic mice without increasing high-density lipoprotein-cholesterol concentrations. Lymphatic vessels are now recognized as prerequisite players in the modulation of cholesterol removal from the artery wall in experimental conditions of plaque regression, and particular attention has been brought to the role of the collecting lymphatic vessels in early atherosclerosis-related lymphatic dysfunction. In the present study, we address whether and how preservation of collecting lymphatic function contributes to the protective effect of apoA-I. METHODS AND RESULTS: Atherosclerotic Ldlr-/- mice treated with low-dose lipid-free apoA-I showed enhanced lymphatic transport and abrogated collecting lymphatic vessel permeability in atherosclerotic Ldlr-/- mice when compared with albumin-control mice. Treatment of human lymphatic endothelial cells with apoA-I increased the adhesion of human platelets on lymphatic endothelial cells, in a bridge-like manner, a mechanism that could strengthen endothelial cell-cell junctions and limit atherosclerosis-associated collecting lymphatic vessel dysfunction. Experiments performed with blood platelets isolated from apoA-I-treated Ldlr-/- mice revealed that apoA-I decreased ex vivo platelet aggregation. This suggests that in vivo apoA-I treatment limits platelet thrombotic potential in blood while maintaining the platelet activity needed to sustain adequate lymphatic function. CONCLUSIONS: Altogether, we bring forward a new pleiotropic role for apoA-I in lymphatic function and unveil new potential therapeutic targets for the prevention and treatment of atherosclerosis.


Sujet(s)
Maladies de l'aorte/prévention et contrôle , Apolipoprotéine A-I/administration et posologie , Athérosclérose/prévention et contrôle , Vaisseaux lymphatiques/effets des médicaments et des substances chimiques , Animaux , Aorte/effets des médicaments et des substances chimiques , Aorte/métabolisme , Aorte/anatomopathologie , Maladies de l'aorte/génétique , Maladies de l'aorte/métabolisme , Maladies de l'aorte/anatomopathologie , Apolipoprotéine A-I/pharmacocinétique , Athérosclérose/génétique , Athérosclérose/métabolisme , Athérosclérose/anatomopathologie , Cellules cultivées , Modèles animaux de maladie humaine , Cellules endothéliales/effets des médicaments et des substances chimiques , Cellules endothéliales/métabolisme , Cellules endothéliales/anatomopathologie , Femelle , Prédisposition génétique à une maladie , Humains , Injections sous-cutanées , Métabolisme lipidique/effets des médicaments et des substances chimiques , Vaisseaux lymphatiques/métabolisme , Vaisseaux lymphatiques/anatomopathologie , Souris de lignée C57BL , Souris knockout , Perméabilité , Phénotype , Plaque d'athérosclérose , Adhésivité plaquettaire/effets des médicaments et des substances chimiques , Agrégation plaquettaire/effets des médicaments et des substances chimiques , Récepteurs aux lipoprotéines LDL/déficit , Récepteurs aux lipoprotéines LDL/génétique , Transduction du signal/effets des médicaments et des substances chimiques , Récepteur-3 au facteur croissance endothéliale vasculaire/métabolisme
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...