Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 25
Filtrer
1.
Nat Med ; 29(4): 859-868, 2023 04.
Article de Anglais | MEDLINE | ID: mdl-36928816

RÉSUMÉ

One of the great challenges in therapeutic oncology is determining who might achieve survival benefits from a particular therapy. Studies on longitudinal circulating tumor DNA (ctDNA) dynamics for the prediction of survival have generally been small or nonrandomized. We assessed ctDNA across 5 time points in 466 non-small-cell lung cancer (NSCLC) patients from the randomized phase 3 IMpower150 study comparing chemotherapy-immune checkpoint inhibitor (chemo-ICI) combinations and used machine learning to jointly model multiple ctDNA metrics to predict overall survival (OS). ctDNA assessments through cycle 3 day 1 of treatment enabled risk stratification of patients with stable disease (hazard ratio (HR) = 3.2 (2.0-5.3), P < 0.001; median 7.1 versus 22.3 months for high- versus low-intermediate risk) and with partial response (HR = 3.3 (1.7-6.4), P < 0.001; median 8.8 versus 28.6 months). The model also identified high-risk patients in an external validation cohort from the randomized phase 3 OAK study of ICI versus chemo in NSCLC (OS HR = 3.73 (1.83-7.60), P = 0.00012). Simulations of clinical trial scenarios employing our ctDNA model suggested that early ctDNA testing outperforms early radiographic imaging for predicting trial outcomes. Overall, measuring ctDNA dynamics during treatment can improve patient risk stratification and may allow early differentiation between competing therapies during clinical trials.


Sujet(s)
Carcinome pulmonaire non à petites cellules , ADN tumoral circulant , Tumeurs du poumon , Humains , Carcinome pulmonaire non à petites cellules/traitement médicamenteux , Carcinome pulmonaire non à petites cellules/génétique , ADN tumoral circulant/génétique , Tumeurs du poumon/traitement médicamenteux , Tumeurs du poumon/génétique , Marqueurs biologiques tumoraux/génétique
2.
J Steroid Biochem Mol Biol ; 190: 115-125, 2019 06.
Article de Anglais | MEDLINE | ID: mdl-30940596

RÉSUMÉ

Serum concentrations of lathosterol, the plant sterols campesterol and sitosterol and the cholesterol metabolite 5α-cholestanol are widely used as surrogate markers of cholesterol synthesis and absorption, respectively. Increasing numbers of laboratories utilize a broad spectrum of well-established and recently developed methods for the determination of cholesterol and non-cholesterol sterols (NCS). In order to evaluate the quality of these measurements and to identify possible sources of analytical errors our group initiated the first international survey for cholesterol and NCS. The cholesterol and NCS survey was structured as a two-part survey which took place in the years 2013 and 2014. The first survey part was designed as descriptive, providing information about the variation of reported results from different laboratories. A set of two lyophilized pooled sera (A and B) was sent to twenty laboratories specialized in chromatographic lipid analysis. The different sterols were quantified either by gas chromatography-flame ionization detection, gas chromatography- or liquid chromatography-mass selective detection. The participants were requested to determine cholesterol and NCS concentrations in the provided samples as part of their normal laboratory routine. The second part was designed as interventional survey. Twenty-two laboratories agreed to participate and received again two different lyophilized pooled sera (C and D). In contrast to the first international survey, each participant received standard stock solutions with defined concentrations of cholesterol and NCS. The participants were requested to use diluted calibration solutions from the provided standard stock solutions for quantification of cholesterol and NCS. In both surveys, each laboratory used its own internal standard (5α-cholestane, epicoprostanol or deuterium labelled sterols). Main outcome of the survey was, that unacceptably high interlaboratory variations for cholesterol and NCS concentrations are reported, even when the individual laboratories used the same calibration material. We discuss different sources of errors and recommend all laboratories analysing cholesterol and NCS to participate in regular quality control programs.


Sujet(s)
Cholestérol/sang , Phytostérols/sang , Dihydrocholestérol/sang , Cholestérol/analogues et dérivés , Chromatographie en phase gazeuse/méthodes , Chromatographie en phase liquide/méthodes , Humains , Sitostérol/sang , Enquêtes et questionnaires
3.
Atherosclerosis ; 277: 256-261, 2018 10.
Article de Anglais | MEDLINE | ID: mdl-30270055

RÉSUMÉ

BACKGROUND AND AIMS: Marked hypercholesterolemia, defined as low density lipoprotein cholesterol (LDL-C) levels ≥ 190 mg/dL, may be due to LDLR, APOB, and PCSK9 variants. In a recent analysis, only 1.7% of cases had such variants. Our goal was to identify other potential genetic causes of hypercholesterolemia. METHODS: In a total of 51,253 subjects with lipid testing, 3.8% had elevated total cholesterol >300 mg/dL and/or LDL-C≥190 mg/dL. Of these, 246 were further studied, and 69 without kidney, liver, or thyroid disease and who met Dutch Lipid Clinic Network criteria of ≥6 points had DNA sequencing done at the LDLR, APOB, PCSK9, APOE, LDLRAP1, STAP1, ABCG5, ABCG8, CYP27A1, LIPA, LIPC, LIPG, LPL, and SCARB1 gene loci and also had 10 SNP analysis for a weighted high LDL-C genetic risk score. RESULTS: In the 69 subjects with genetic analyses, the following variants were observed in 37 subjects (53.6%): LDLR (n = 20, 2 novel), ABCG5/8 (n = 7, 2 novel), APOB (n = 3, 1 novel), CYP27A1 (n = 3, 1 novel), LIPA (n = 2, 1 novel), APOE (n = 2), LIPC (n = 1, novel), LIPG (n = 1, novel), and SCARB1 (n = 1); 14 subjects (20.3%) had a high polygenic score, with 4 (5.8%) having no variants. CONCLUSIONS: Our data indicate that in addition to variants in LDLR, APOB, PCSK9, APOE, LDLRAP1, and STAP1, variants in ABCG5/8, CYP27A1, LIPA, LIPC, and LIPG may be associated with hypercholesterolemia and such information should be used to optimize therapy.


Sujet(s)
Cholestérol LDL/sang , Variation génétique , Hyperlipoprotéinémie de type II/génétique , Argentine/épidémiologie , Marqueurs biologiques/sang , Bases de données factuelles , Femelle , Prédisposition génétique à une maladie , Dépistage génétique , Humains , Hyperlipoprotéinémie de type II/sang , Hyperlipoprotéinémie de type II/diagnostic , Hyperlipoprotéinémie de type II/épidémiologie , Mâle , Adulte d'âge moyen , Phénotype , Prévalence , Pronostic , Appréciation des risques , Facteurs de risque
4.
J Lipid Res ; 59(12): 2421-2435, 2018 12.
Article de Anglais | MEDLINE | ID: mdl-30333156

RÉSUMÉ

We assessed secondary and genetic causes of severe HDL deficiency in 258,252 subjects, of whom 370 men (0.33%) and 144 women (0.099%) had HDL cholesterol levels <20 mg/dl. We excluded 206 subjects (40.1%) with significant elevations of triglycerides, C-reactive protein, glycosylated hemoglobin, myeloperoxidase, or liver enzymes and men receiving testosterone. We sequenced 23 lipid-related genes in 201 (65.3%) of 308 eligible subjects. Mutations (23 novel) and selected variants were found at the following gene loci: 1) ABCA1 (26.9%): 2 homozygotes, 7 compound or double heterozygotes, 30 heterozygotes, and 2 homozygotes and 13 heterozygotes with variants rs9282541/p.R230C or rs111292742/c.-279C>G; 2) LCAT (12.4%): 1 homozygote, 3 compound heterozygotes, 13 heterozygotes, and 8 heterozygotes with variant rs4986970/p.S232T; 3) APOA1 (5.0%): 1 homozygote and 9 heterozygotes; and 4) LPL (4.5%): 1 heterozygote and 8 heterozygotes with variant rs268/p.N318S. In addition, 4.5% had other mutations, and 46.8% had no mutations. Atherosclerotic cardiovascular disease (ASCVD) prevalence rates in the ABCA1, LCAT, APOA1, LPL, and mutation-negative groups were 37.0%, 4.0%, 40.0%, 11.1%, and 6.4%, respectively. Severe HDL deficiency is uncommon, with 40.1% having secondary causes and 48.8% of the subjects sequenced having ABCA1, LCAT, APOA1, or LPL mutations or variants, with the highest ASCVD prevalence rates being observed in the ABCA1 and APOA1 groups.


Sujet(s)
Maladies cardiovasculaires/étiologie , Maladies cardiovasculaires/génétique , Hypoalphalipoprotéinémies/étiologie , Hypoalphalipoprotéinémies/génétique , Membre-1 de la sous-famille A des transporteurs à cassette liant l'ATP/génétique , Apolipoprotéine A-I/génétique , Protéine C-réactive/métabolisme , Cholestérol HDL/génétique , Femelle , Hétérozygote , Homozygote , Humains , Lipoprotéines HDL/génétique , Mâle , Mutation/génétique
5.
J Clin Lipidol ; 12(1): 152-161, 2018.
Article de Anglais | MEDLINE | ID: mdl-29169939

RÉSUMÉ

BACKGROUND: Sitosterolemia is associated with increases in intestinal sterol absorption, low-density lipoprotein cholesterol (LDL-C), and cardiovascular disease risk. OBJECTIVE: We examined the relationship between hypercholesterolemia and sitosterolemia in a large population and report a new sitosterolemia case. METHODS: Plasma sterol concentrations were measured by gas chromatography/mass spectrometry, and LDL-C by direct assay. RESULTS: Of 207,926 subjects tested, 4.3% had LDL-C ≥190 mg/dL. Plasma ß-sitosterol concentrations ≥8.0 mg/L (99th percentile) were found in 4.3% of these subjects vs 0.72% with LDL-C <130 mg/dL. Among all subjects, 0.050% had ß-sitosterol levels ≥15.0 mg/L, consistent with sitosterolemia, while among those with LDL-C ≥190 mg/dL, 0.334% had this rare disorder. A 13-year-old boy with the highest LDL-C (679 mg/dL) of all subjects had planar xanthomas and a ß-sitosterol level of 53.5 mg/L (normal <3.3 mg/L). He was a compound heterozygote for 2 ABCG8 mutations (p.N409D and an intron 11+2T>A splice site mutation). On a low-cholesterol and plant-sterol diet, his LDL-C decreased to 485 mg/dL (-29%) and ß-sitosterol to 44.6 mg/L (-27%). On atorvastatin 20 mg/d, his LDL-C decreased to 299 mg/dL (-38%). With added ezetimibe 10 mg/d, his LDL-C normalized to 60 mg/dL (-80% further decrease); and his ß-sitosterol decreased to 14.1 mg/L (-68% further decrease). CONCLUSIONS: Our data indicate that about 4% of subjects with LDL-C concentrations ≥190 mg/dL have plasma ß-sitosterol concentrations above the 99th percentile and about 0.3% have concentrations consistent with sitosterolemia. Therefore, this diagnosis should be considered in such patients.


Sujet(s)
Hypercholestérolémie/diagnostic , Maladies intestinales/diagnostic , Erreurs innées du métabolisme lipidique/diagnostic , Phytostérols/effets indésirables , Membre-8 de la sous-famille G des transporteurs à cassette liant l'ATP/génétique , Adolescent , Adulte , Sujet âgé , Anticholestérolémiants/usage thérapeutique , Cholestérol LDL/sang , Régime pauvre en graisses , Ézétimibe/usage thérapeutique , Femelle , Hétérozygote , Humains , Hypercholestérolémie/sang , Hypercholestérolémie/complications , Hypercholestérolémie/traitement médicamenteux , Maladies intestinales/sang , Maladies intestinales/complications , Erreurs innées du métabolisme lipidique/sang , Erreurs innées du métabolisme lipidique/complications , Mâle , Adulte d'âge moyen , Phytostérols/sang , Sitostérol/sang
6.
Prog Cardiovasc Dis ; 59(2): 97-106, 2016.
Article de Anglais | MEDLINE | ID: mdl-27565770

RÉSUMÉ

Low serum high density lipoprotein cholesterol level (HDL-C) <40 mg/dL in men and <50 mg/dL in women is a significant independent risk factor for cardiovascular disease (CVD), and is often observed in patients with hypertriglyceridemia, obesity, insulin resistance, and diabetes. Patients with marked deficiency of HDL-C (<20 mg/dL) in the absence of secondary causes are much less common (<1% of the population). These patients may have homozygous, compound heterozygous, or heterozygous defects involving the apolipoprotein (APO)AI, ABCA1, or lecithin:cholesterol acyl transferase genes, associated with apo A-I deficiency, apoA-I variants, Tangier disease , familial lecithin:cholesteryl ester acyltransferase deficiency, and fish eye disease. There is marked variability in laboratory and clinical presentation, and DNA analysis is necessary for diagnosis. These patients can develop premature CVD, neuropathy, kidney failure, neuropathy, hepatosplenomegaly and anemia. Treatment should be directed at optimizing all non-HDL risk factors.


Sujet(s)
Hypoalphalipoprotéinémies , Prise en charge de la maladie , Humains , Hypoalphalipoprotéinémies/diagnostic , Hypoalphalipoprotéinémies/étiologie , Hypoalphalipoprotéinémies/thérapie
7.
J Clin Lipidol ; 9(3): 390-5, 2015.
Article de Anglais | MEDLINE | ID: mdl-26073399

RÉSUMÉ

We report a novel heterozygous apolipoprotein A-I (apoA-I) missense mutation (c.517C>A, p.Arg149Ser, designated as apoA-IBoston) in a 67-year-old woman and her 2 sons, who had mean serum high-density lipoprotein (HDL) cholesterol, apoA-I, and apoA-I in very large α-1 HDL that were 10%, 35%, and 16% of normal, respectively (all P < .05). The percentage of HDL cholesterol in the esterified form was also significantly (P < .05) reduced to 52% of control values. Cholesteryl ester tranfer protein (CETP) activity was normal. The mean global, adenosine triphosphate (ATP)-binding cassette transporter A1 and scavenger receptor B type I-mediated cellular cholesterol efflux capacity in apoB-depleted serum from affected family members were 41%, 37%, 47%, 54%, and 48% of control values, respectively (all P < .05). lecithin-cholesterol acyltransferase (LCAT) activity in plasma was 71% of controls, whereas in the cell-based assay, it was 73% of control values (P < .05). The data indicate that this novel apoA-I missense is associated with markedly decreased levels of HDL cholesterol and very large α-1 HDL, as well as decreased serum cellular cholesterol efflux and LCAT activity, but not with premature coronary heart disease, similar to other apoA-I mutations that have been associated with decreased LCAT activity.


Sujet(s)
Apolipoprotéine A-I/génétique , Cholestérol/métabolisme , Mutation faux-sens , Phosphatidylcholine-Sterol O-Acyltransferase/métabolisme , Adulte , Sujet âgé , Substitution d'acide aminé , Apolipoprotéine A-I/métabolisme , Activation enzymatique/génétique , Femelle , Humains , Mâle , Phosphatidylcholine-Sterol O-Acyltransferase/génétique
8.
Atherosclerosis ; 235(2): 470-6, 2014 Aug.
Article de Anglais | MEDLINE | ID: mdl-24950002

RÉSUMÉ

OBJECTIVE: We report a novel apolipoprotein (apo) A-I truncation (apoA-IMytilene) due to a heterozygous nonsense mutation (c.718C > T, p.Gln216*) in a 68-year-old male proband with premature coronary heart disease (CHD), corneal arcus, and very low plasma concentrations of HDL cholesterol (HDL-C) and apoA-I. Two family members also had the same mutation. Our objectives were to characterize the kindred and to examine the kinetics of apoA-I, as well as cellular cholesterol efflux capacity in the proband. METHODS: We carried out the kinetic studies using a primed constant infusion of [5,5,5-D3]L-leucine and isotopic enrichment was determined by gas chromatography mass spectrometry in the proband and seven controls with low HDL-C. To assess cellular cholesterol efflux capacity, we used a validated ex vivo system that involved incubation of J774 macrophages with apoB-depleted serum from the proband, five controls with normal HDL-C, and two controls with low HDL-C. RESULTS: Stable isotope kinetic studies indicated that the proband had an apoA-I production rate (PR) that was 41% lower than the mean PR observed in low HDL-C controls (n = 7). The cellular cholesterol efflux capacity assessment showed normalized cholesterol efflux capacity in the proband was decreased by 36% compared to the mean normalized cholesterol efflux capacity of normal controls (n = 5). CONCLUSIONS: Our data indicate that this novel heterozygous apoA-I truncation is associated with markedly decreased levels of HDL-C, plasma apoA-I, and apoA-I in large α-1 HDL particles, as well as decreased total cellular cholesterol efflux and decreased apoA-I production.


Sujet(s)
Apolipoprotéine A-I/génétique , Cholestérol HDL/sang , Maladie coronarienne/sang , Adulte , Sujet âgé , Apolipoprotéine A-I/biosynthèse , Apolipoprotéine A-I/sang , Codon non-sens , Deutérium , Humains , Cinétique , Leucine/métabolisme , Lipoprotéine (a)/métabolisme , Mâle , Adulte d'âge moyen , Pedigree
9.
Atherosclerosis ; 235(1): 176-81, 2014 Jul.
Article de Anglais | MEDLINE | ID: mdl-24854628

RÉSUMÉ

AIMS: Our goals were to examine the relationships of a specific ATP-binding cassette transporter A1 (ABCA1) variant, rs2230806 (R219K), on baseline lipids, low-density lipoprotein cholesterol (LDL-C) lowering due to pravastatin, baseline heart disease, and cardiac endpoints on trial. METHODS AND RESULTS: The ABCA1 R219K variant was assessed in 5414 participants in PROSPER (PROspective Study of Pravastatin in the Elderly at Risk) (mean age 75.3 years), who had been randomized to pravastatin 40 mg/day or placebo and followed for a mean of 3.2 years. Of these subjects 47.6% carried the variant, with 40.0% carrying one allele, and 7.6% carrying both alleles. No effects on baseline LDL-C levels were noted, but mean HDL-C increased modestly according to the number of variant alleles being present (1.27 vs 1.28 vs 1.30 mmol/L, p = 0.024). No relationships between the presence or absence of this variant and statin induced LDL-C lowering response or CHD at baseline were noted. However within trial those with the variant as compared to those without the variant, the overall adjusted hazard ratio for new cardiovascular disease (fatal CHD, non-fatal myocardial infarction, or fatal or non-fatal stroke) was 1.22 (95% CI 1.06-1.40, p = 0.006), while for those in the pravastatin group it was 1.41 (1.15-1.73, p = 0.001), and for those in the placebo group it was 1.08 (0.89-1.30, p = 0.447) (p for interaction 0.058). CONCLUSION: Our data indicate that subjects with the ABCA1 R219K variant may get significantly less heart disease risk reduction from pravastatin treatment than those without the variant.


Sujet(s)
Membre-1 de la sous-famille A des transporteurs à cassette liant l'ATP/génétique , Cardiopathies/traitement médicamenteux , Polymorphisme de nucléotide simple , Pravastatine/usage thérapeutique , Sujet âgé , Allèles , Cholestérol HDL/sang , Cholestérol LDL/sang , Cholestérol LDL/métabolisme , Maladie coronarienne/sang , Maladie coronarienne/traitement médicamenteux , Maladie coronarienne/mortalité , Femelle , Génotype , Cardiopathies/sang , Cardiopathies/mortalité , Humains , Inhibiteurs de l'hydroxyméthylglutaryl-CoA réductase/usage thérapeutique , Mâle , Infarctus du myocarde/sang , Infarctus du myocarde/traitement médicamenteux , Modèles des risques proportionnels , Accident vasculaire cérébral/sang , Accident vasculaire cérébral/traitement médicamenteux
10.
J Clin Lipidol ; 7(1): 82-7, 2013.
Article de Anglais | MEDLINE | ID: mdl-23351586

RÉSUMÉ

Tangier disease is a rare autosomal-recessive disorder caused by mutation in the ATP binding cassette transporter 1 (ABCA1) gene. Typically, Tangier disease manifests with symptoms and signs resulting from the deposition of cholesteryl esters in nonadipose tissues; chiefly, in peripheral nerves leading to neuropathy and in reticulo-endothelial organs, such as liver, spleen, lymph nodes, and tonsils, causing their enlargement and discoloration. An association with early cardiovascular disease can be variable. We describe a patient with a unique phenotype of Tangier disease from a novel splice site mutation in the ABCA1 gene that is associated with a central nervous system presentation resembling multiple sclerosis, and the presence of premature atherosclerosis.


Sujet(s)
Transporteurs ABC/génétique , Maladie de Tangier/génétique , Membre-1 de la sous-famille A des transporteurs à cassette liant l'ATP , Transporteurs ABC/métabolisme , Adulte , Cholestérol HDL/sang , Cholestérol LDL/sang , Coloscopie , Stroma de la cornée/métabolisme , Stroma de la cornée/anatomopathologie , Analyse de mutations d'ADN , Femelle , Humains , Imagerie par résonance magnétique , Muscles squelettiques/anatomopathologie , Mutation , Pedigree , Phénotype , Maladie de Tangier/diagnostic , Tomodensitométrie
11.
Atherosclerosis ; 220(2): 413-7, 2012 Feb.
Article de Anglais | MEDLINE | ID: mdl-22189199

RÉSUMÉ

Our goal was to determine whether genetic variation at genes affecting statin metabolism or targets of statin therapy would influence low density lipoprotein (LDL) cholesterol lowering with pravastatin, baseline heart disease, or cardiac endpoints on trial. We examined associations of single nucleotide polymorphisms (SNPs) at the liver X receptor alpha (LXRA, rs12221497), and the solute carrier organic anion transporter (SLCO1B1, rs4149056 and rs2306283) gene loci with these variables. We studied 5411 participants in PROSPER (PROspective Study of Pravastatin in the Elderly at Risk) (mean age 75.3 years), who had been randomized to pravastatin 40 mg/day or placebo and were followed for a mean of 3.2 years. No relationships between genetic variation at the LXRA gene locus with statin induced LDL lowering response or other parameters were noted. Both the SLCO1B1 rs4149056 (valine for alanine at 174) and the rs2306283 (asparagine for aspartic acid at 130) SNPs affect the amino acid sequence of the SLCO1B1 gene product. No effect of the rs2306283 SNP on any of the variables was noted. However the presence of the rs4149056 SNP was associated with significantly less LDL cholesterol lowering response to pravastatin (wildtype, 71.5% of the population, -37.0%; heterozygotes, 25.8% of the population, -36.0%; and homozygotes, 2.7% of the population, -31.8%, p=0.003 at 6 months, and p=0.022 at 12 months). Our data indicate that the presence of the rs4149056 non-synonymous SNP at the SLCO1B1 gene locus can significantly decrease the pravastatin induced LDL cholesterol lowering response.


Sujet(s)
Cholestérol LDL/sang , Inhibiteurs de l'hydroxyméthylglutaryl-CoA réductase/usage thérapeutique , Hypercholestérolémie/traitement médicamenteux , Hypercholestérolémie/génétique , Transporteurs d'anions organiques/génétique , Polymorphisme de nucléotide simple , Pravastatine/usage thérapeutique , Facteurs âges , Sujet âgé , Sujet âgé de 80 ans ou plus , Analyse de variance , Marqueurs biologiques/sang , Maladies cardiovasculaires/sang , Maladies cardiovasculaires/génétique , Maladies cardiovasculaires/prévention et contrôle , Loi du khi-deux , Europe , Femelle , Hétérozygote , Homozygote , Humains , Hypercholestérolémie/sang , Hypercholestérolémie/complications , Déséquilibre de liaison , Récepteurs hépatiques X , Polypeptide C de transport d'anions organiques , Modèles logistiques , Mâle , Récepteurs nucléaires orphelins/génétique , Phénotype , Études prospectives , Appréciation des risques , Facteurs de risque , Facteurs temps , Résultat thérapeutique
12.
Atherosclerosis ; 220(2): 456-62, 2012 Feb.
Article de Anglais | MEDLINE | ID: mdl-22192511

RÉSUMÉ

Single nucleotide polymorphisms (SNPs) at the KIF6 (kinesin like protein 6, rs20455 or 719Arg), LPA (lipoprotein(a), rs3798220), TAS2R50 (taste receptor type 2, member 50, rs1376251) and VAMP8 (vesicle-associated membrane protein 8, rs1010) have previously been associated with low density lipoprotein cholesterol (LDL-C) lowering response to statins, coronary heart disease (CHD) at baseline, or CHD events on trial. We examined SNPs at the KIF6 (rs20455 or 719Arg), LPA (rs3798220), TAS2R50 (rs1376251) and VAMP8 (rs1010) in 5,411 participants in PROSPER (PROspective Study of Pravastatin in the Elderly at Risk) (mean age 75.3 years), who had been randomized to pravastatin 40 mg/day or placebo and were followed for a mean of 3.2 years. No SNP was related to vascular disease at baseline. Only the KIF6 SNP was related to LDL-C lowering with homozygous Arg 719 subjects being significantly less responsive than other groups (p=0.025, -34.2 vs. -36.1%). With regard to the primary CHD endpoint on trial (fatal or non-fatal myocardial infarction or stroke), we observed a significant relationship for KIF6 719Arg homozygotes (p=0.03, hazards ratio 0.47, 12.8% of the population) in women on pravastatin only, and for TAS2R50 for the AA genotype (p=0.03, hazards ratio 1.76, 8.9% of the population), also only in women on pravastatin. Our data indicate that the assessment of KIF6 rs20455 and TAS2R50 rs1376251 genotypes are not useful for predicting statin induced cardiovascular risk reduction in men, but do predict CHD risk reduction in women in this elderly population. However, these differences are no longer significant after correction for multiple comparisons, and we do not recommend the assessment of any of these SNPs in clinical practice.


Sujet(s)
Maladies cardiovasculaires/génétique , Maladies cardiovasculaires/prévention et contrôle , Cholestérol LDL/sang , Inhibiteurs de l'hydroxyméthylglutaryl-CoA réductase/usage thérapeutique , Hypercholestérolémie/traitement médicamenteux , Hypercholestérolémie/génétique , Kinésine/génétique , Lipoprotéine (a)/génétique , Pravastatine/usage thérapeutique , Protéines R-SNARE/génétique , Récepteurs couplés aux protéines G/génétique , Facteurs âges , Sujet âgé , Sujet âgé de 80 ans ou plus , Analyse de variance , Maladies cardiovasculaires/sang , Loi du khi-deux , Europe , Femelle , Homozygote , Humains , Hypercholestérolémie/sang , Hypercholestérolémie/complications , Déséquilibre de liaison , Mâle , Phénotype , Polymorphisme de nucléotide simple , Études prospectives , Appréciation des risques , Facteurs de risque , Facteurs temps , Résultat thérapeutique
13.
J Clin Lipidol ; 5(6): 493-9, 2011.
Article de Anglais | MEDLINE | ID: mdl-22108153

RÉSUMÉ

BACKGROUND: A case of homozygous familial lecithin:cholesterol acyltransferase (LCAT) deficiency with a novel homozygous LCAT missense mutation (replacement of methionine by arginine at position 293 in the amino acid sequence of the LCAT protein) is reported. METHODS AND RESULTS: The probable diagnosis was suggested by findings of marked high density lipoprotein (HDL) deficiency, corneal opacification, anemia, and renal insufficiency. The diagnosis was confirmed by two dimensional gel electrophoresis of HDL, the measurement of free and esterified cholesterol, and sequencing of the LCAT gene. CONCLUSIONS: In our view the most important aspects of therapy to prevent the kidney disease that these patients develop is careful control of blood pressure and lifestyle measures to optimize non HDL lipoproteins. In the future replacement therapy by gene transfer or other methods may become available.


Sujet(s)
Homozygote , Déficit en lécithine-cholestérol-acyl-transférase/génétique , Mutation faux-sens , Phosphatidylcholine-Sterol O-Acyltransferase/métabolisme , Séquence d'acides aminés , Apolipoprotéine A-I/sang , Cholestérol/sang , Opacité cornéenne/génétique , Opacité cornéenne/métabolisme , Électrophorèse bidimensionnelle sur gel , Humains , Immunotransfert , Déficit en lécithine-cholestérol-acyl-transférase/sang , Déficit en lécithine-cholestérol-acyl-transférase/métabolisme , Lipoprotéines HDL/sang , Mâle , Adulte d'âge moyen , Phosphatidylcholine-Sterol O-Acyltransferase/sang , Phosphatidylcholine-Sterol O-Acyltransferase/génétique , Protéinurie/génétique , Protéinurie/métabolisme , Analyse de séquence d'ADN
14.
PLoS One ; 6(9): e23890, 2011.
Article de Anglais | MEDLINE | ID: mdl-21915265

RÉSUMÉ

BACKGROUND: Plasma concentrations of C-reactive protein (CRP), a marker of chronic inflammation, have been associated with cognitive impairment in old age. However, it is unknown whether CRP is causally linked to cognitive decline. METHODS AND FINDINGS: Within the Prospective Study of Pravastatin in the Elderly at Risk (PROSPER) trial, with 5680 participants with a mean age of 75 years, we examined associations of CRP levels and its genetic determinants with cognitive performance and decline over 3.2 years mean follow-up. Higher plasma CRP concentrations were associated with poorer baseline performance on the Stroop test (P = 0.001) and Letter Digit Tests (P<0.001), but not with the immediate and delayed Picture Learning Test (PLT; both P>0.5). In the prospective analyses, higher CRP concentrations associated with increased rate of decline in the immediate PLT (P = 0.016), but not in other cognitive tests (all p>0.11). Adjustment for prevalent cardiovascular risk factors and disease did not change the baseline associations nor associations with cognitive decline during follow-up. Four haplotypes of CRP were used and, compared to the common haplotype, carrierships associated strongly with levels of CRP (all P<0.007). In comparison to strong associations of apolipoprotein E with cognitive measures, associations of CRP haplotypes with such measures were inconsistent. CONCLUSION: Plasma CRP concentrations associate with cognitive performance in part through pathways independent of (risk factors for) cardiovascular disease. However, lifelong exposure to higher CRP levels does not associate with poorer cognitive performance in old age. The current data weaken the argument for a causal role of CRP in cognitive performance, but further study is warranted to draw definitive conclusions.


Sujet(s)
Protéine C-réactive/génétique , Protéine C-réactive/métabolisme , Troubles de la cognition/sang , Troubles de la cognition/génétique , Sujet âgé , Sujet âgé de 80 ans ou plus , Vieillissement/génétique , Vieillissement/physiologie , Femelle , Génotype , Humains , Mâle , Tests neuropsychologiques , Polymorphisme génétique/génétique
15.
J Lipid Res ; 51(5): 1201-7, 2010 May.
Article de Anglais | MEDLINE | ID: mdl-19752398

RÉSUMÉ

Niemann-Pick C1-like 1 protein (NPC1L1) plays a critical role in intestinal cholesterol absorption. Our objective was to examine whether five variants (-133A>G, -18A>C, L272L, V1296V, and U3_28650A>G) at the NPC1L1 gene have effects on lipid levels, prevalence, and incidence of coronary heart disease (CHD) and lipid-lowering response to pravastatin. We studied 5,804 elderly participants from the PROSPER study, who were randomized to prava-statin 40 mg/day or placebo and were followed on average for 3.2 years. In the adjusted gender-pooled analyses, homozygous carriers of the minor alleles at four NPC1L1 sites (-18A>C, L272L, V1296V, and U3_28650A>G, minor allele frequencies 0.15-0.33) had 2-8% higher LDL-cholesterol (LDL-C) levels at baseline than homozygous carriers of the common alleles (P < 0.05). Homozygotes for the rare alleles also had a significant increase in the risk of CHD events on trial (range of hazard ratios 1.50-1.67; P < 0.02), regardless of the treatment regimen. The -133 A>G polymorphism and not other variants was associated with 6 month LDL-C lowering (P = 0.02). Our data indicate that variation in the NPC1L1 gene is associated with plasma total and LDL-C levels and CHD risk.


Sujet(s)
Maladie coronarienne/sang , Maladie coronarienne/génétique , Locus génétiques/génétique , Prédisposition génétique à une maladie , Variation génétique , Lipoprotéines/sang , Protéines membranaires/génétique , Sujet âgé , Sujet âgé de 80 ans ou plus , Maladie coronarienne/traitement médicamenteux , Maladie coronarienne/épidémiologie , Femelle , Humains , Mâle , Protéines de transport membranaire , Polymorphisme de nucléotide simple , Pravastatine/usage thérapeutique , Risque , Résultat thérapeutique
16.
J Clin Endocrinol Metab ; 95(1): 93-9, 2010 Jan.
Article de Anglais | MEDLINE | ID: mdl-19906786

RÉSUMÉ

CONTEXT: Associations between adiposity and circulating inflammation markers are assumed to be causal, although the direction of the relationship has not been proven. OBJECTIVE: The aim of the study was to explore the causal direction of the relationship between adiposity and inflammation using a bidirectional Mendelian randomization approach. METHODS: In the PROSPER study of 5804 elderly patients, we related C-reactive protein (CRP) single nucleotide polymorphisms (SNPs) (rs1800947 and rs1205) and adiposity SNPs (FTO and MC4R) to body mass index (BMI) as well as circulating levels of CRP and leptin. We gave each individual two allele scores ranging from zero to 4, counting each pair of alleles related to CRP levels or BMI. RESULTS: With increasing CRP allele score, there was a stepwise decrease in CRP levels (P for trend < 0.0001) and a 1.98 mg/liter difference between extremes of the allele score distribution, but there was no associated change in BMI or leptin levels (P >or= 0.89). By contrast, adiposity allele score was associated with 1) an increase in BMI (1.2 kg/m(2) difference between extremes; P for trend 0.002); 2) an increase in circulating leptin (5.77 ng/ml difference between extremes; P for trend 0.0027); and 3) increased CRP levels (1.24 mg/liter difference between extremes; P for trend 0.002). CONCLUSIONS: Greater adiposity conferred by FTO and MC4R SNPs led to higher CRP levels, with no evidence for any reverse pathway. Future studies should extend our findings to other circulating inflammatory parameters. This study illustrates the potential power of Mendelian randomization to dissect directions of causality between intercorrelated metabolic factors.


Sujet(s)
Adiposité/génétique , Liaison génétique , Inflammation/génétique , Analyse de randomisation mendélienne/méthodes , Sujet âgé , Sujet âgé de 80 ans ou plus , Alpha-ketoglutarate-dependent dioxygenase FTO , Marqueurs biologiques/sang , Protéine C-réactive/génétique , Femelle , Marqueurs génétiques/physiologie , Génotype , Humains , Inflammation/sang , Mâle , Polymorphisme de nucléotide simple , Protéines/génétique , Récepteur de la mélanocortine de type 4/génétique
18.
Atherosclerosis ; 200(1): 109-14, 2008 Sep.
Article de Anglais | MEDLINE | ID: mdl-18261733

RÉSUMÉ

Our purpose was to evaluate associations of single nucleotide polymorphisms (SNPs) at the low density lipoprotein (LDL) receptor (LDLR C44857T, minor allele frequency (MAF) 0.26, and A44964G, MAF 0.25, both in the untranslated region) and HMG-CoA reductase (HMGCR i18 T>G, MAF 0.019) gene loci with baseline lipid values, statin-induced LDL-cholesterol (C) lowering response, and incident coronary heart disease (CHD) and cardiovascular disease (CVD) on trial. Our population consisted of 5804 elderly men and women with vascular disease or one or more vascular disease risk factors, who were randomly allocated to pravastatin or placebo. Other risk factors and apolipoprotein (apo) E phenotype were controlled for in the analysis. Despite a prior report, no relationships with the HMGCR SNP were noted. For the LDLR SNPs C44857T and A44964G we noted significant associations of the rare alleles with baseline LDL-C and triglyceride levels, a modest association of the C44857T with LDL-C lowering to pravastatin in men, and significant associations with incident CHD and CVD of both SNPs, especially in men on pravastatin. Our data indicate that genetic variation at the LDLR locus can affect baseline lipids, response to pravastatin, and CVD risk in subjects placed on statin treatment.


Sujet(s)
Prédisposition génétique à une maladie/génétique , Hydroxymethylglutaryl-CoA reductases/génétique , Inhibiteurs de l'hydroxyméthylglutaryl-CoA réductase/pharmacologie , Polymorphisme de nucléotide simple/génétique , Pravastatine/pharmacologie , Récepteurs aux lipoprotéines LDL/génétique , Sujet âgé , Sujet âgé de 80 ans ou plus , Maladies cardiovasculaires/génétique , Maladies cardiovasculaires/prévention et contrôle , Cholestérol HDL/effets des médicaments et des substances chimiques , Cholestérol LDL/effets des médicaments et des substances chimiques , Femelle , Humains , Mâle , Facteurs de risque , Triglycéride
19.
Atherosclerosis ; 200(1): 95-101, 2008 Sep.
Article de Anglais | MEDLINE | ID: mdl-18262190

RÉSUMÉ

Caucasian carriers of the T allele at R46L in the proprotein convertase subtilisin/kexin type 9 (PCSK9) locus have been reported to have 15% lower low-density lipoprotein (LDL) cholesterol (C) levels and 47% lower coronary heart disease (CHD) risk. Our objective was to examine two PCSK9 single nucleotide polymorphisms (SNPs), R46L and E670G, in 5783 elderly participants in Prospective Study of Pravastatin in the Elderly at Risk (PROSPER), of whom 43% had a history of vascular disease at baseline, and who were randomized to pravastatin or placebo with followup. In this population 3.5% were carriers of the T allele at R46L, and these subjects had significantly (p<0.001) lower levels of LDL C (mean, -10%), no difference in LDL C lowering response to pravastatin, and a non-significant 19% unadjusted and 9% adjusted decreased risk of vascular disease at baseline, with no on trial effect. Moreover, 6.0% were carriers of the G allele at E670G with no significant relationships with baseline LDL C, response to pravastatin, or vascular disease risk being observed. Our data support the concept that the rare allele of the R46L SNP at the PCSK9 locus significantly lowers LDL C, but does not greatly reduce CHD risk in an elderly population with a high prevalence of cardiovascular disease.


Sujet(s)
Cholestérol LDL/génétique , Maladie coronarienne/génétique , Inhibiteurs de l'hydroxyméthylglutaryl-CoA réductase , Polymorphisme de nucléotide simple/génétique , Pravastatine , Serine endopeptidases/génétique , Sujet âgé , Sujet âgé de 80 ans ou plus , Cholestérol LDL/effets des médicaments et des substances chimiques , Maladie coronarienne/traitement médicamenteux , Femelle , Prédisposition génétique à une maladie/génétique , Humains , Inhibiteurs de l'hydroxyméthylglutaryl-CoA réductase/pharmacologie , Déséquilibre de liaison , Mâle , Odds ratio , Pravastatine/génétique , Pravastatine/pharmacologie , Proprotéine convertase 9 , Proprotein convertases
20.
J Lipid Res ; 49(2): 349-57, 2008 Feb.
Article de Anglais | MEDLINE | ID: mdl-17991756

RÉSUMÉ

Our aim was to characterize HDL subspecies and fat-soluble vitamin levels in a kindred with familial apolipoprotein A-I (apoA-I) deficiency. Sequencing of the APOA1 gene revealed a nonsense mutation at codon -2, Q[-2]X, with two documented homozygotes, eight heterozygotes, and two normal subjects in the kindred. Homozygotes presented markedly decreased HDL cholesterol levels, undetectable plasma apoA-1, tuboeruptive and planar xanthomas, mild corneal arcus and opacification, and severe premature coronary artery disease. In both homozygotes, analysis of HDL particles by two-dimensional gel electrophoresis revealed undetectable apoA-I, decreased amounts of small alpha-3 migrating apoA-II particles, and only modestly decreased normal amounts of slow alpha migrating apoA-IV- and apoE-containing HDL, while in the eight heterozygotes, there was loss of large alpha-1 HDL particles. There were no significant decreases in plasma fat-soluble vitamin levels noted in either homozygotes or heterozygotes compared with normal control subjects. Our data indicate that isolated apoA-I deficiency results in marked HDL deficiency with very low apoA-II alpha-3 HDL particles, modest reductions in the separate and distinct plasma apoA-IV and apoE HDL particles, tuboeruptive xanthomas, premature coronary atherosclerosis, and no evidence of fat malabsorption.


Sujet(s)
Apolipoprotéine A-I/déficit , Apolipoprotéine A-I/génétique , Hypolipoprotéinémies/génétique , Hypolipoprotéinémies/métabolisme , Lipoprotéines HDL/composition chimique , Adulte , Sujet âgé , Apolipoprotéine A-I/sang , Enfant , Enfant d'âge préscolaire , Cholestérol HDL/sang , Femelle , Humains , Hypolipoprotéinémies/sang , Lipoprotéines HDL/sang , Mâle , Taille de particule , Pedigree , Xanthomatose/métabolisme
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE