Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
1.
J Clin Lipidol ; 14(4): 470-481, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32620384

RESUMO

BACKGROUND: Genetic factors partly determine the risk for premature myocardial infarction (MI). OBJECTIVES: We report the identification of a novel rare genetic variant in a kindred with an autosomal dominant trait for premature MI and atherosclerosis and explored the association of a common nonsynonymous variant in the same gene with the risk of ischemic heart disease (IHD) in a population-based study. METHODS: Next-generation sequencing was performed in a small pedigree with premature MI or subclinical atherosclerosis. A common variant, rs8141797 A>G (p.Asn466Ser), in sushi domain-containing protein 2 (SUSD2) was studied in the prospective Copenhagen General Population Studies (N = 105,408) for association with IHD. RESULTS: A novel heterozygous nonsense mutation in SUSD2 (c.G583T; p.Glu195Ter) was associated with the disease phenotype in the pedigree. SUSD2 protein was expressed in aortic specimens in the subendothelial cell layer and around the vasa vasorum. Furthermore, the minor G-allele of rs8141797 was associated with per allele higher levels of SUSD2 mRNA expression in the heart and vasculature. In the Copenhagen General Population Study, hazard ratios for IHD were 0.92 (95% CI: 0.87-0.97) in AG heterozygotes and 0.86 (0.62-1.19) in GG homozygotes vs noncarrriers (P-trend = .002). Finally, in meta-analysis including 73,983 IHD cases and 215,730 controls, the odds ratio for IHD per G-allele vs A-allele was 0.93 (0.90-0.96) (P = 4.6 × 10-7). CONCLUSIONS: The identification of a truncating mutation in SUSD2, which was associated with premature MI and subclinical atherosclerosis, combined with the finding that a common missense variant in SUSD2 was strongly associated with a lower risk of IHD, suggest that SUSD2 may alter the risk of atherosclerosis.


Assuntos
Predisposição Genética para Doença/genética , Glicoproteínas de Membrana/genética , Isquemia Miocárdica/genética , Adulto , Idoso , Estudos de Casos e Controles , Códon sem Sentido , Feminino , Heterozigoto , Homozigoto , Humanos , Masculino , Metanálise como Assunto , Pessoa de Meia-Idade
2.
J Clin Lipidol ; 12(6): 1493-1501.e11, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30249512

RESUMO

BACKGROUND: Plasma apolipoprotein C-III (apoC-III) levels are associated with coronary artery disease (CAD) risk. OBJECTIVE: To assess whether lipoprotein-associated apoC-III levels predict risk of CAD events. METHODS: apoC-III associated with apoB, apoAI, and Lp(a) (apoCIII-apoB, apoCIII-apoAI, and apoCIII-Lp(a), respectively) were measured using high-throughput chemiluminescent enzyme-linked immunoassays in 2711 subjects (1879 controls and 832 cases with CAD) in the European Prospective Investigation into Cancer and Nutrition-Norfolk prospective population study with 7.4 years of follow-up. These measures were correlated with a variety of lipid measurements and the presence of CAD. The indices of "total apoCIII-apoB" and "total apoCIII-apoAI" were derived by multiplying plasma apoB and apoAI, respectively. RESULTS: apoCIII-apoB (P = .001), apoCIII-Lp(a) (P < .001), apoCIII-apoAI (P = .005) were higher in cases vs controls; tended to correlate positively with body mass index, hsCRP, apoC-III, low-density lipoprotein (LDL) cholesterol, triglycerides, remnant cholesterol, very low density lipoprotein, LDL and high-density lipoprotein particle number and very low density lipoprotein size; but negatively with LDL and high-density lipoprotein particle size (P < .001 for all). apoCIII-apoB, apoCIII-apoAI, apoCIII-Lp(a), total apoCIII-Lp(a), and total apoCIII-apoB were predictors of CAD after adjustment of age, sex, body mass index, smoking, diabetes, hypertensive and lipid-lowering drug use, but they lost their significance after further adjustment of lipid and lipoprotein variables. CONCLUSIONS: This study suggests that enzyme-linked immunoassay-measured lipoprotein-associated apoC-III markers reflect atherogenic lipid particles but do not independently predict risk of CAD events.


Assuntos
Apolipoproteína C-III/sangue , Doença da Artéria Coronariana/sangue , Idoso , Apolipoproteína A-I/sangue , Apolipoproteína B-100/sangue , Feminino , Seguimentos , Humanos , Masculino , Pessoa de Meia-Idade , Estudos Prospectivos , Fatores de Risco
3.
Arterioscler Thromb Vasc Biol ; 38(7): 1440-1453, 2018 07.
Artigo em Inglês | MEDLINE | ID: mdl-29853565

RESUMO

OBJECTIVE: Studies into the role of LRP1 (low-density lipoprotein receptor-related protein 1) in human lipid metabolism are scarce. Although it is known that a common variant in LRP1 (rs116133520) is significantly associated with HDL-C (high-density lipoprotein cholesterol), the mechanism underlying this observation is unclear. In this study, we set out to study the functional effects of 2 rare LRP1 variants identified in subjects with extremely low HDL-C levels. APPROACH AND RESULTS: In 2 subjects with HDL-C below the first percentile for age and sex and moderately elevated triglycerides, we identified 2 rare variants in LRP1: p.Val3244Ile and p.Glu3983Asp. Both variants decrease LRP1 expression and stability. We show in a series of translational experiments that these variants culminate in reduced trafficking of ABCA1 (ATP-binding cassette A1) to the cell membrane. This is accompanied by an increase in cell surface expression of SR-B1 (scavenger receptor class B type 1). Combined these effects may contribute to low HDL-C levels in our study subjects. Supporting these findings, we provide epidemiological evidence that rs116133520 is associated with apo (apolipoprotein) A1 but not with apoB levels. CONCLUSIONS: This study provides the first evidence that rare variants in LRP1 are associated with changes in human lipid metabolism. Specifically, this study shows that LRP1 may affect HDL metabolism by virtue of its effect on both ABCA1 and SR-B1.


Assuntos
Transportador 1 de Cassete de Ligação de ATP/metabolismo , HDL-Colesterol/metabolismo , Fibroblastos/metabolismo , Variação Genética , Hipoalfalipoproteinemias/sangue , Hipoalfalipoproteinemias/genética , Proteína-1 Relacionada a Receptor de Lipoproteína de Baixa Densidade/genética , Receptores Depuradores Classe B/metabolismo , Apolipoproteína A-I/sangue , Linhagem Celular Tumoral , Membrana Celular/metabolismo , Predisposição Genética para Doença , Células HEK293 , Humanos , Hipoalfalipoproteinemias/diagnóstico , Fígado/metabolismo , Proteína-1 Relacionada a Receptor de Lipoproteína de Baixa Densidade/metabolismo , Fenótipo , Estudos Prospectivos , Estabilidade Proteica , Transporte Proteico , Triglicerídeos/sangue
4.
Arterioscler Thromb Vasc Biol ; 37(11): 2147-2155, 2017 11.
Artigo em Inglês | MEDLINE | ID: mdl-28882873

RESUMO

OBJECTIVE: High-density lipoproteins (HDL) are considered to protect against atherosclerosis in part by facilitating the removal of cholesterol from peripheral tissues. However, factors regulating lipid efflux are incompletely understood. We previously identified a variant in adenosine triphosphate-binding cassette transporter A8 (ABCA8) in an individual with low HDL cholesterol (HDLc). Here, we investigate the role of ABCA8 in cholesterol efflux and in regulating HDLc levels. APPROACH AND RESULTS: We sequenced ABCA8 in individuals with low and high HDLc and identified, exclusively in low HDLc probands, 3 predicted deleterious heterozygous ABCA8 mutations (p.Pro609Arg [P609R], IVS17-2 A>G and p.Thr741Stop [T741X]). HDLc levels were lower in heterozygous mutation carriers compared with first-degree family controls (0.86±0.34 versus 1.17±0.26 mmol/L; P=0.005). HDLc levels were significantly decreased by 29% (P=0.01) in Abca8b-/- mice on a high-cholesterol diet compared with wild-type mice, whereas hepatic overexpression of human ABCA8 in mice resulted in significant increases in plasma HDLc and the first steps of macrophage-to-feces reverse cholesterol transport. Overexpression of wild-type but not mutant ABCA8 resulted in a significant increase (1.8-fold; P=0.01) of cholesterol efflux to apolipoprotein AI in vitro. ABCA8 colocalizes and interacts with adenosine triphosphate-binding cassette transporter A1 and further potentiates adenosine triphosphate-binding cassette transporter A1-mediated cholesterol efflux. CONCLUSIONS: ABCA8 facilitates cholesterol efflux and modulates HDLc levels in humans and mice.


Assuntos
Transportadores de Cassetes de Ligação de ATP/metabolismo , Colesterol na Dieta/sangue , HDL-Colesterol/sangue , Transportadores de Cassetes de Ligação de ATP/deficiência , Transportadores de Cassetes de Ligação de ATP/genética , Adulto , Idoso , Animais , Apolipoproteína A-I/sangue , Apolipoproteína B-100/sangue , Transporte Biológico , Biomarcadores/sangue , Células COS , Estudos de Casos e Controles , Chlorocebus aethiops , Análise Mutacional de DNA , Dieta Hiperlipídica , Fezes/química , Feminino , Células HEK293 , Hereditariedade , Heterozigoto , Humanos , Fígado/metabolismo , Macrófagos/metabolismo , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Pessoa de Meia-Idade , Mutação , Linhagem , Fenótipo , Transfecção
5.
J Am Heart Assoc ; 6(8)2017 Aug 03.
Artigo em Inglês | MEDLINE | ID: mdl-28775061

RESUMO

BACKGROUND: The contribution of apolipoprotein A-I (apoA-I) to coronary heart disease (CHD) risk stratification over and above high-density lipoprotein cholesterol (HDL-C) is unclear. We studied the associations between plasma levels of HDL-C and apoA-I, either alone or combined, with risk of CHD events and cardiovascular risk factors among apparently healthy men and women. METHODS AND RESULTS: HDL-C and apoA-I levels were measured among 17 661 participants of the EPIC (European Prospective Investigation into Cancer)-Norfolk prospective population study. Hazard ratios for CHD events and distributions of risk factors were calculated by quartiles of HDL-C and apoA-I. Results were validated using data from the ARIC (Atherosclerosis Risk in Communities) and WHS (Women's Health Study) cohorts, comprising 15 494 and 27 552 individuals, respectively. In EPIC-Norfolk, both HDL-C and apoA-I quartiles were strongly and inversely associated with CHD risk. Within HDL-C quartiles, higher apoA-I levels were not associated with lower CHD risk; in fact, CHD risk was higher within some HDL-C quartiles. ApoA-I levels were associated with higher levels of CHD risk factors: higher body mass index, HbA1c, non-HDL-C, triglycerides, apolipoprotein B, systolic blood pressure, and C-reactive protein, within fixed HDL-C quartiles. In contrast, HDL-C levels were consistently inversely associated with overall CHD risk and CHD risk factors within apoA-I quartiles (P<0.001). These findings were validated in the ARIC and WHS cohorts. CONCLUSIONS: Our findings demonstrate that apoA-I levels do not offer predictive information over and above HDL-C. In fact, within some HDL-C quartiles, higher apoA-I levels were associated with higher risk of CHD events, possibly because of the unexpected higher prevalence of cardiovascular risk factors in association with higher apoA-I levels. CLINICAL TRIAL REGISTRATION: URL: https://www.clinicaltrials.gov. Unique identifier: NCT00000479.


Assuntos
Apolipoproteína A-I/metabolismo , HDL-Colesterol/metabolismo , Doença das Coronárias/metabolismo , Saúde da Mulher , Idoso , Antineoplásicos/administração & dosagem , Aspirina/administração & dosagem , Doenças Cardiovasculares/prevenção & controle , Doença das Coronárias/mortalidade , Método Duplo-Cego , Inglaterra/epidemiologia , Feminino , Humanos , Estimativa de Kaplan-Meier , Masculino , Pessoa de Meia-Idade , Neoplasias/prevenção & controle , Inibidores da Agregação Plaquetária/administração & dosagem , Prognóstico , Estudos Prospectivos , Fatores de Risco , Vitamina E/administração & dosagem
6.
Arterioscler Thromb Vasc Biol ; 37(6): 1206-1212, 2017 06.
Artigo em Inglês | MEDLINE | ID: mdl-28473441

RESUMO

OBJECTIVE: Apolipoprotein C-III (apoC-III) is a key regulator of triglyceride metabolism. Elevated triglyceride-rich lipoproteins and apoC-III levels are causally linked to coronary artery disease (CAD) risk. The mechanism(s) through which apoC-III increases CAD risk remains largely unknown. The aim was to confirm the association between apoC-III plasma levels and CAD risk and to explore which lipoprotein subfractions contribute to this relationship between apoC-III and CAD risk. APPROACH AND RESULTS: Plasma apoC-III levels were measured in baseline samples from a nested case-control study in the European Prospective Investigation of Cancer (EPIC)-Norfolk study. The study comprised 2711 apparently healthy study participants, of whom 832 subsequently developed CAD. We studied the association of baseline apoC-III levels with incident CAD risk, lipoprotein subfractions measured by nuclear magnetic resonance spectroscopy and inflammatory biomarkers. ApoC-III levels were significantly associated with CAD risk (odds ratio, 1.91; 95% confidence interval, 1.48-2.48 for highest compared with lowest quintile), retaining significance after adjustment for traditional CAD risk factors (odds ratio, 1.47; 95% confidence interval, 1.11-1.94). ApoC-III levels were positively correlated with triglyceride levels, (r=0.39), particle numbers of very-low-density lipoprotein (r=0.25), intermediate-density lipoprotein (r=0.23), small dense low-density lipoprotein (r=0.26), and high-sensitivity C-reactive protein (r=0.15), whereas an inverse correlation was observed with large low-density lipoprotein particle number (r=-0.11), P<0.001 for each. Mediation analysis indicated that the association between apoC-III and CAD risk could be explained by triglyceride elevation (triglyceride, very-low-density lipoprotein, and intermediate-density lipoprotein particles), small low-density lipoprotein particle size, and high-sensitivity C-reactive protein. CONCLUSIONS: ApoC-III levels are significantly associated with incident CAD risk. Elevated levels of remnant lipoproteins, small dense low-density lipoprotein, and low-grade inflammation may explain this association.


Assuntos
Apolipoproteína C-III/sangue , Doença da Artéria Coronariana/sangue , Doença da Artéria Coronariana/epidemiologia , Idoso , Biomarcadores/sangue , Proteína C-Reativa/análise , Estudos de Casos e Controles , Doença da Artéria Coronariana/diagnóstico , Inglaterra/epidemiologia , Feminino , Humanos , Incidência , Mediadores da Inflamação/sangue , Lipoproteínas LDL/sangue , Modelos Logísticos , Masculino , Pessoa de Meia-Idade , Análise Multivariada , Ressonância Magnética Nuclear Biomolecular , Razão de Chances , Tamanho da Partícula , Prognóstico , Estudos Prospectivos , Medição de Risco , Fatores de Risco , Fatores de Tempo , Triglicerídeos/sangue , Regulação para Cima
7.
J Am Coll Cardiol ; 69(7): 823-836, 2017 Feb 21.
Artigo em Inglês | MEDLINE | ID: mdl-28209224

RESUMO

BACKGROUND: Genome-wide association studies have so far identified 56 loci associated with risk of coronary artery disease (CAD). Many CAD loci show pleiotropy; that is, they are also associated with other diseases or traits. OBJECTIVES: This study sought to systematically test if genetic variants identified for non-CAD diseases/traits also associate with CAD and to undertake a comprehensive analysis of the extent of pleiotropy of all CAD loci. METHODS: In discovery analyses involving 42,335 CAD cases and 78,240 control subjects we tested the association of 29,383 common (minor allele frequency >5%) single nucleotide polymorphisms available on the exome array, which included a substantial proportion of known or suspected single nucleotide polymorphisms associated with common diseases or traits as of 2011. Suggestive association signals were replicated in an additional 30,533 cases and 42,530 control subjects. To evaluate pleiotropy, we tested CAD loci for association with cardiovascular risk factors (lipid traits, blood pressure phenotypes, body mass index, diabetes, and smoking behavior), as well as with other diseases/traits through interrogation of currently available genome-wide association study catalogs. RESULTS: We identified 6 new loci associated with CAD at genome-wide significance: on 2q37 (KCNJ13-GIGYF2), 6p21 (C2), 11p15 (MRVI1-CTR9), 12q13 (LRP1), 12q24 (SCARB1), and 16q13 (CETP). Risk allele frequencies ranged from 0.15 to 0.86, and odds ratio per copy of the risk allele ranged from 1.04 to 1.09. Of 62 new and known CAD loci, 24 (38.7%) showed statistical association with a traditional cardiovascular risk factor, with some showing multiple associations, and 29 (47%) showed associations at p < 1 × 10-4 with a range of other diseases/traits. CONCLUSIONS: We identified 6 loci associated with CAD at genome-wide significance. Several CAD loci show substantial pleiotropy, which may help us understand the mechanisms by which these loci affect CAD risk.


Assuntos
Doença da Artéria Coronariana/genética , Loci Gênicos , Pleiotropia Genética , Estudos de Casos e Controles , Doença da Artéria Coronariana/epidemiologia , Feminino , Frequência do Gene , Estudo de Associação Genômica Ampla , Humanos , Masculino , Razão de Chances , Polimorfismo de Nucleotídeo Único
8.
J Am Soc Nephrol ; 28(4): 1278-1285, 2017 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-27799487

RESUMO

CKD associates with a 1.5- to 3.5-fold increased risk for cardiovascular disease. Both diseases are characterized by increased inflammation, and in patients with CKD, elevated C-reactive protein level predicts cardiovascular risk. In addition to systemic inflammation, local arterial inflammation, driven by monocyte-derived macrophages, predicts future cardiovascular events in the general population. We hypothesized that subjects with CKD have increased arterial and cellular inflammation, reflected by 18F-fluorodeoxyglucose (18F-FDG) positron emission tomography computed tomography (PET/CT) of the arterial wall and a migratory phenotype of monocytes. We assessed 18F-FDG uptake in the arterial wall in 14 patients with CKD (mean±SD age: 59±5 years, mean±SD eGFR: 37±12 ml/min per 1.73 m2) but without cardiovascular diseases, diabetes, or inflammatory conditions and in 14 control subjects (mean age: 60±11 years, mean eGFR: 86±16 ml/min per 1.73 m2). Compared with controls, patients with CKD showed increased arterial inflammation, quantified as target-to-background ratio (TBR) in the aorta (TBRmax: CKD, 3.14±0.70 versus control, 2.12±0.27; P=0.001) and the carotid arteries (TBRmax: CKD, 2.45±0.65 versus control, 1.66±0.27; P<0.001). Characterization of circulating monocytes using flow cytometry revealed increased chemokine receptor expression and enhanced transendothelial migration capacity in patients with CKD compared with controls. In conclusion, this increased arterial wall inflammation, observed in patients with CKD but without overt atherosclerotic disease and with few traditional risk factors, may contribute to the increased cardiovascular risk associated with CKD. The concomitant elevation of monocyte activity may provide novel therapeutic targets for attenuating this inflammation and thereby preventing CKD-associated cardiovascular disease.


Assuntos
Arterite/diagnóstico por imagem , Arterite/etiologia , Doenças Cardiovasculares/diagnóstico por imagem , Doenças Cardiovasculares/etiologia , Fluordesoxiglucose F18 , Inflamação/diagnóstico por imagem , Inflamação/etiologia , Tomografia por Emissão de Pósitrons combinada à Tomografia Computadorizada , Compostos Radiofarmacêuticos , Insuficiência Renal Crônica/complicações , Células , Estudos Transversais , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Fatores de Risco
9.
J Clin Lipidol ; 10(5): 1137-1144.e3, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27678430

RESUMO

BACKGROUND: TA-8995 is a potent inhibitor of cholesteryl ester transfer protein (CETP) with beneficial effects on lipids and lipoproteins. The effect of TA-8995 on cholesterol efflux capacity (CEC), a measure of high-density lipoprotein (HDL) function, and HDL subparticle distribution is largely unknown. OBJECTIVE: To assess the effect of the CETP inhibitor TA-8995 on ABCA1- and non-ABCA1-driven CEC and on HDL particle distribution. METHODS: Total, non-ABCA1-, and ABCA1-specific CEC from J774 cells and HDL subclass distribution assessed by two-dimensional gel electrophoresis were measured at baseline and after 12-week treatment in 187 mild-dyslipidemic patients randomized to placebo, 1 mg, 5 mg, 10 mg TA-8995, or 10 mg TA-8995 combined with 10 mg rosuvastatin (NCT01970215). RESULTS: Compared with placebo, total, non-ABCA1-, and ABCA1-specific CEC were increased dose dependently by up to 38%, 72%, and 28%, respectively, in patients randomized to 10 mg of TA-8995. PreBeta-1 HDL, the primary acceptor for ABCA1-driven cholesterol efflux, was increased by 36%. This increase in preBeta-1 HDL correlated significantly with the total and the ABCA1-driven CEC increase, whereas the high-density lipoprotein cholesterol (HDL-C) increase did not. CONCLUSION: TA-8995 dose dependently increased not only total and non-ABCA1-specific CEC but also ABCA1-specific CEC and preBeta-1 HDL particle levels. These findings suggest that TA-8995 not only increases HDL-C levels but also promotes functional properties of HDL particles. This CETP inhibitor-driven preBeta-1 HDL increase is an important predictor of both ABCA1 and total CEC increase, independent of HDL-C increase. Whether these changes in HDL particle composition and functionality have a beneficial effect on cardiovascular outcome requires formal testing in a cardiovascular outcome trial.


Assuntos
Anticolesterolemiantes/uso terapêutico , Proteínas de Transferência de Ésteres de Colesterol/antagonistas & inibidores , Dislipidemias/tratamento farmacológico , Lipoproteínas HDL/análise , Quinolinas/uso terapêutico , Transportador 1 de Cassete de Ligação de ATP/metabolismo , Adolescente , Adulto , Idoso , Linhagem Celular , Proteínas de Transferência de Ésteres de Colesterol/metabolismo , HDL-Colesterol/análise , HDL-Colesterol/sangue , LDL-Colesterol/sangue , Método Duplo-Cego , Quimioterapia Combinada , Feminino , Lipoproteínas de Alta Densidade Pré-beta/análise , Humanos , Lipoproteínas HDL/classificação , Masculino , Pessoa de Meia-Idade , Efeito Placebo , Rosuvastatina Cálcica/uso terapêutico , Triglicerídeos/sangue , Adulto Jovem
10.
Lancet ; 388(10057): 2239-2253, 2016 11 05.
Artigo em Inglês | MEDLINE | ID: mdl-27665230

RESUMO

BACKGROUND: Elevated lipoprotein(a) (Lp[a]) is a highly prevalent (around 20% of people) genetic risk factor for cardiovascular disease and calcific aortic valve stenosis, but no approved specific therapy exists to substantially lower Lp(a) concentrations. We aimed to assess the efficacy, safety, and tolerability of two unique antisense oligonucleotides designed to lower Lp(a) concentrations. METHODS: We did two randomised, double-blind, placebo-controlled trials. In a phase 2 trial (done in 13 study centres in Canada, the Netherlands, Germany, Denmark, and the UK), we assessed the effect of IONIS-APO(a)Rx, an oligonucleotide targeting apolipoprotein(a). Participants with elevated Lp(a) concentrations (125-437 nmol/L in cohort A; ≥438 nmol/L in cohort B) were randomly assigned (in a 1:1 ratio in cohort A and in a 4:1 ratio in cohort B) with an interactive response system to escalating-dose subcutaneous IONIS-APO(a)Rx (100 mg, 200 mg, and then 300 mg, once a week for 4 weeks each) or injections of saline placebo, once a week, for 12 weeks. Primary endpoints were mean percentage change in fasting plasma Lp(a) concentration at day 85 or 99 in the per-protocol population (participants who received more than six doses of study drug) and safety and tolerability in the safety population. In a phase 1/2a first-in-man trial, we assessed the effect of IONIS-APO(a)-LRx, a ligand-conjugated antisense oligonucleotide designed to be highly and selectively taken up by hepatocytes, at the BioPharma Services phase 1 unit (Toronto, ON, Canada). Healthy volunteers (Lp[a] ≥75 nmol/L) were randomly assigned to receive a single dose of 10-120 mg IONIS-APO(a)LRx subcutaneously in an ascending-dose design or placebo (in a 3:1 ratio; single-ascending-dose phase), or multiple doses of 10 mg, 20 mg, or 40 mg IONIS-APO(a)LRx subcutaneously in an ascending-dose design or placebo (in an 8:2 ratio) at day 1, 3, 5, 8, 15, and 22 (multiple-ascending-dose phase). Primary endpoints were mean percentage change in fasting plasma Lp(a) concentration, safety, and tolerability at day 30 in the single-ascending-dose phase and day 36 in the multiple-ascending-dose phase in participants who were randomised and received at least one dose of study drug. In both trials, the randomised allocation sequence was generated by Ionis Biometrics or external vendor with a permuted-block randomisation method. Participants, investigators, sponsor personnel, and clinical research organisation staff who analysed the data were all masked to the treatment assignments. Both trials are registered with ClinicalTrials.gov, numbers NCT02160899 and NCT02414594. FINDINGS: From June 25, 2014, to Nov 18, 2015, we enrolled 64 participants to the phase 2 trial (51 in cohort A and 13 in cohort B). 35 were randomly assigned to IONIS-APO(a)Rx and 29 to placebo. At day 85/99, participants assigned to IONIS-APO(a)Rx had mean Lp(a) reductions of 66·8% (SD 20·6) in cohort A and 71·6% (13·0) in cohort B (both p<0·0001 vs pooled placebo). From April 15, 2015, to Jan 11, 2016, we enrolled 58 healthy volunteers to the phase 1/2a trial of IONIS-APO(a)-LRx. Of 28 participants in the single-ascending-dose phase, three were randomly assigned to 10 mg, three to 20 mg, three to 40 mg, six to 80 mg, six to 120 mg, and seven to placebo. Of 30 participants in the multiple-ascending-dose phase, eight were randomly assigned to 10 mg, eight to 20 mg, eight to 40 mg, and six to placebo. Significant dose-dependent reductions in mean Lp(a) concentrations were noted in all single-dose IONIS-APO(a)-LRx groups at day 30. In the multidose groups, IONIS-APO(a)-LRx resulted in mean reductions in Lp(a) of 66% (SD 21·8) in the 10 mg group, 80% (SD 13·7%) in the 20 mg group, and 92% (6·5) in the 40 mg group (p=0·0007 for all vs placebo) at day 36. Both antisense oligonucleotides were safe. There were two serious adverse events (myocardial infarctions) in the IONIS-APO(a)Rx phase 2 trial, one in the IONIS-APO(a)Rx and one in the placebo group, but neither were thought to be treatment related. 12% of injections with IONIS-APO(a)Rx were associated with injection-site reactions. IONIS-APO(a)-LRx was associated with no injection-site reactions. INTERPRETATION: IONIS-APO(a)-LRx is a novel, tolerable, potent therapy to reduce Lp(a) concentrations. IONIS-APO(a)-LRx might mitigate Lp(a)-mediated cardiovascular risk and is being developed for patients with elevated Lp(a) concentrations with existing cardiovascular disease or calcific aortic valve stenosis. FUNDING: Ionis Pharmaceuticals.


Assuntos
Apolipoproteínas A/administração & dosagem , Apoproteína(a)/antagonistas & inibidores , Lipoproteína(a) , Oligonucleotídeos Antissenso/administração & dosagem , Apolipoproteínas A/genética , Doenças Cardiovasculares/tratamento farmacológico , Método Duplo-Cego , Feminino , Humanos , Lipoproteína(a)/sangue , Lipoproteína(a)/genética , Masculino , Pessoa de Meia-Idade , Fatores de Risco , Resultado do Tratamento
11.
Atherosclerosis ; 249: 140-7, 2016 06.
Artigo em Inglês | MEDLINE | ID: mdl-27105157

RESUMO

BACKGROUND AND AIMS: Among subjects with high-density-lipoprotein cholesterol (HDL-C) below the 1st percentile in the general population, we identified a heterozygous variant OSBPL1A p.C39X encoding a short truncated protein fragment that co-segregated with low plasma HDL-C. METHODS: We investigated the composition and function of HDL from the carriers and non-carriers and studied the properties of the mutant protein in cultured hepatocytes. RESULTS: Plasma HDL-C and apolipoprotein (apo) A-I were lower in carriers versus non-carriers, whereas the other analyzed plasma components or HDL parameters did not differ. Sera of the carriers displayed a reduced capacity to act as cholesterol efflux acceptors (p < 0.01), whereas the cholesterol acceptor capacity of their isolated HDL was normal. Fibroblasts from a p.C39X carrier showed reduced cholesterol efflux to lipid-free apoA-I but not to mature HDL particles, suggesting a specific defect in ABCA1-mediated efflux pathway. In hepatic cells, GFP-OSBPL1A partially co-localized in endosomes containing fluorescent apoA-I, suggesting that OSBPL1A may regulate the intracellular handling of apoA-I. The GFP-OSBPL1A-39X mutant protein remained in the cytosol and failed to interact with Rab7, which normally recruits OSBPL1A to late endosomes/lysosomes, suggesting that this mutation represents a loss-of-function. CONCLUSIONS: The present work represents the first characterization of a human OSBPL1A mutation. Our observations provide evidence that a familial loss-of-function mutation in OSBPL1A affects the first step of the reverse cholesterol transport process and associates with a low HDL-C phenotype. This suggests that rare mutations in OSBPL genes may contribute to dyslipidemias.


Assuntos
Apolipoproteína A-I/metabolismo , HDL-Colesterol/sangue , Receptores de Esteroides/genética , Adulto , Idoso , Animais , Carcinoma Hepatocelular/metabolismo , Linhagem Celular , Citosol/metabolismo , Dislipidemias/genética , Endossomos/metabolismo , Feminino , Fibroblastos/metabolismo , Proteínas de Fluorescência Verde/metabolismo , Heterozigoto , Humanos , Lisossomos/metabolismo , Masculino , Camundongos , Pessoa de Meia-Idade , Mutação , Linhagem , Fenótipo , Células RAW 264.7 , Reprodutibilidade dos Testes , Proteínas rab de Ligação ao GTP/metabolismo , proteínas de unión al GTP Rab7
12.
N Engl J Med ; 374(12): 1134-44, 2016 03 24.
Artigo em Inglês | MEDLINE | ID: mdl-26934567

RESUMO

BACKGROUND: The discovery of low-frequency coding variants affecting the risk of coronary artery disease has facilitated the identification of therapeutic targets. METHODS: Through DNA genotyping, we tested 54,003 coding-sequence variants covering 13,715 human genes in up to 72,868 patients with coronary artery disease and 120,770 controls who did not have coronary artery disease. Through DNA sequencing, we studied the effects of loss-of-function mutations in selected genes. RESULTS: We confirmed previously observed significant associations between coronary artery disease and low-frequency missense variants in the genes LPA and PCSK9. We also found significant associations between coronary artery disease and low-frequency missense variants in the genes SVEP1 (p.D2702G; minor-allele frequency, 3.60%; odds ratio for disease, 1.14; P=4.2×10(-10)) and ANGPTL4 (p.E40K; minor-allele frequency, 2.01%; odds ratio, 0.86; P=4.0×10(-8)), which encodes angiopoietin-like 4. Through sequencing of ANGPTL4, we identified 9 carriers of loss-of-function mutations among 6924 patients with myocardial infarction, as compared with 19 carriers among 6834 controls (odds ratio, 0.47; P=0.04); carriers of ANGPTL4 loss-of-function alleles had triglyceride levels that were 35% lower than the levels among persons who did not carry a loss-of-function allele (P=0.003). ANGPTL4 inhibits lipoprotein lipase; we therefore searched for mutations in LPL and identified a loss-of-function variant that was associated with an increased risk of coronary artery disease (p.D36N; minor-allele frequency, 1.9%; odds ratio, 1.13; P=2.0×10(-4)) and a gain-of-function variant that was associated with protection from coronary artery disease (p.S447*; minor-allele frequency, 9.9%; odds ratio, 0.94; P=2.5×10(-7)). CONCLUSIONS: We found that carriers of loss-of-function mutations in ANGPTL4 had triglyceride levels that were lower than those among noncarriers; these mutations were also associated with protection from coronary artery disease. (Funded by the National Institutes of Health and others.).


Assuntos
Angiopoietinas/genética , Moléculas de Adesão Celular/genética , Doença da Artéria Coronariana/genética , Lipase Lipoproteica/genética , Mutação , Triglicerídeos/sangue , Idoso , Proteína 4 Semelhante a Angiopoietina , Feminino , Técnicas de Genotipagem , Humanos , Lipase Lipoproteica/antagonistas & inibidores , Lipase Lipoproteica/metabolismo , Masculino , Pessoa de Meia-Idade , Mutação de Sentido Incorreto , Fatores de Risco , Análise de Sequência de DNA , Triglicerídeos/genética
13.
J Lipid Res ; 57(9): 1612-8, 2016 09.
Artigo em Inglês | MEDLINE | ID: mdl-26637277

RESUMO

Lipoprotein (a) [Lp(a)] is a human plasma lipoprotein with unique structural and functional characteristics. Lp(a) is an assembly of two components: a central core with apoB and an additional glycoprotein, called apo(a). Ever since the strong association between elevated levels of Lp(a) and an increased risk for CVD was recognized, interest in the therapeutic modulation of Lp(a) levels has increased. Here, the past and present therapies aiming to lower Lp(a) levels will be reviewed, demonstrating that these agents have had varying degrees of success. The next challenge will be to prove that Lp(a) lowering also leads to cardiovascular benefit in patients with elevated Lp(a) levels. Therefore, highly specific and potent Lp(a)-lowering strategies are awaited urgently.


Assuntos
Apolipoproteína B-100/sangue , Apolipoproteínas A/sangue , Doenças Cardiovasculares/tratamento farmacológico , Lipoproteína(a)/sangue , Doenças Cardiovasculares/sangue , Humanos , Hipolipemiantes/uso terapêutico , Lipoproteína(a)/efeitos dos fármacos , Fatores de Risco
14.
Eur J Hum Genet ; 24(1): 86-91, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25898923

RESUMO

Cardiovascular disease (CVD) is a major cause of death in Western societies. CVD risk is largely genetically determined. The molecular pathology is, however, not elucidated in a large number of families suffering from CVD. We applied exclusion linkage analysis and next-generation sequencing to elucidate the molecular defect underlying premature CVD in a small pedigree, comprising two generations of which six members suffered from premature CVD. A total of three variants showed co-segregation with the disease status in the family. Two of these variants were excluded from further analysis based on the prevalence in replication cohorts, whereas a non-synonymous variant in MCF.2 Cell Line Derived Transforming Sequence-like protein (MCF2L, c.2066A>G; p.(Asp689Gly); NM_001112732.1), located in the DH domain, was only present in the studied family. MCF2L is a guanine exchange factor that potentially links pathways that signal through Rac1 and RhoA. Indeed, in HeLa cells, MCF2L689Gly failed to activate Rac1 as well as RhoA, resulting in impaired stress fiber formation. Moreover, MCF2L protein was found in human atherosclerotic lesions but not in healthy tissue segments. In conclusion, a rare functional variant in MCF2L, leading to impaired DH function, was identified in a small pedigree with premature CVD. The presence of MCF2L in human atherosclerotic plaque specimen lends further support to its potential role in atherosclerosis.


Assuntos
Aterosclerose/genética , Mutação , Placa Aterosclerótica/genética , Fatores de Troca de Nucleotídeo Guanina Rho/genética , Adulto , Idade de Início , Aterosclerose/metabolismo , Aterosclerose/patologia , Sequência de Bases , Estudos de Coortes , Feminino , Regulação da Expressão Gênica , Ligação Genética , Células HeLa , Humanos , Masculino , Pessoa de Meia-Idade , Dados de Sequência Molecular , Linhagem , Placa Aterosclerótica/metabolismo , Placa Aterosclerótica/patologia , Plasmídeos/química , Plasmídeos/genética , Ligação Proteica , Fatores de Troca de Nucleotídeo Guanina Rho/metabolismo , Transdução de Sinais , Transfecção , Proteínas rac1 de Ligação ao GTP/genética , Proteínas rac1 de Ligação ao GTP/metabolismo , Proteína rhoA de Ligação ao GTP/genética , Proteína rhoA de Ligação ao GTP/metabolismo
15.
J Clin Lipidol ; 9(3): 396-9, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26073400

RESUMO

In this report, we present a patient who suffered from a myocardial infarction at an extremely young age. The only remarkable finding in the risk factor workup was a near undetectable high-density lipoprotein (HDL)-cholesterol plasma level (0.09 mmol/L). Genetic analysis of key genes involved in HDL metabolism resulted in the discovery of 2 very rare mutations in the ABCA1 and APOA1 genes. We discuss the effects of these mutations on HDL metabolism and reverse cholesterol transport and interpret these findings in relation to the extensive atherosclerosis at a very young age in this patient.


Assuntos
Transportador 1 de Cassete de Ligação de ATP/deficiência , Apolipoproteína A-I/deficiência , Lipoproteínas HDL/metabolismo , Mutação , Infarto do Miocárdio/genética , Infarto do Miocárdio/metabolismo , Adulto , Humanos , Masculino
16.
Curr Opin Lipidol ; 25(6): 418-22, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25255477

RESUMO

PURPOSE OF REVIEW: Recent large Mendelian randomization studies associate loss-of-function mutations in apolipoprotein CIII (APOCIII) with low levels of triglycerides and decreased incidence of cardiovascular disease. With ample in-vitro and in-vivo evidence for a role of apoCIII in lipoprotein lipase-mediated triglyceride clearance and remnant removal, it is, thus, an attractive target for the treatment of hypertriglyceridemia and the prevention of cardiovascular disease. This review evaluates the current position of apoCIII in clinical practice and provides a glimpse into the future in terms of treatment options. RECENT FINDINGS: Two large Mendelian randomization studies have shown three identical loss-of-function mutations in APOCIII to be linked to favorable lipid profiles and lower incidence of coronary artery disease. A second-generation antisense oligonucleotide, which selectively inhibits apoCIII, was able to decrease serum apoCIII and triglyceride levels in rodents, nonhuman primates and humans. SUMMARY: The central role of apoCIII in hypertriglyceridemia and cardiovascular disease was further cemented by recent findings and promising intervention data that showed the possibility of using antisense therapy to lower apoCIII and triglyceride levels. Currently, planned phase 3 trials should provide answers in regards to long-term efficacy and safety of this novel therapy.


Assuntos
Apolipoproteína C-III/antagonistas & inibidores , Doença da Artéria Coronariana/prevenção & controle , Hipertrigliceridemia/terapia , Oligonucleotídeos Antissenso/genética , Triglicerídeos/antagonistas & inibidores , Animais , Apolipoproteína C-III/sangue , Apolipoproteína C-III/genética , Ensaios Clínicos Fase III como Assunto , Doença da Artéria Coronariana/sangue , Doença da Artéria Coronariana/etiologia , Expressão Gênica , Terapia Genética/métodos , Haplorrinos , Humanos , Hipertrigliceridemia/sangue , Hipertrigliceridemia/complicações , Hipertrigliceridemia/genética , Camundongos , Oligonucleotídeos Antissenso/metabolismo , Pró-Proteína Convertase 9 , Pró-Proteína Convertases/antagonistas & inibidores , Pró-Proteína Convertases/genética , Ensaios Clínicos Controlados Aleatórios como Assunto , Serina Endopeptidases/genética , Triglicerídeos/sangue
17.
PLoS One ; 9(5): e98289, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24879339

RESUMO

AIMS: Genetic factors explain a proportion of the inter-individual variation in the risk for atherosclerotic events, but the genetic basis of atherosclerosis and atherothrombosis in families with Mendelian forms of premature atherosclerosis is incompletely understood. We set out to unravel the molecular pathology in a large kindred with an autosomal dominant inherited form of premature atherosclerosis. METHODS AND RESULTS: Parametric linkage analysis was performed in a pedigree comprising 4 generations, of which a total of 11 members suffered from premature vascular events. A parametric LOD-score of 3.31 was observed for a 4.4 Mb interval on chromosome 12. Upon sequencing, a non-synonymous variant in KERA (c.920C>G; p.Ser307Cys) was identified. The variant was absent from nearly 28,000 individuals, including 2,571 patients with premature atherosclerosis. KERA, a proteoglycan protein, was expressed in lipid-rich areas of human atherosclerotic lesions, but not in healthy arterial specimens. Moreover, KERA expression in plaques was significantly associated with plaque size in a carotid-collar Apoe-/- mice (r2 = 0.69; p<0.0001). CONCLUSION: A rare variant in KERA was identified in a large kindred with premature atherosclerosis. The identification of KERA in atherosclerotic plaque specimen in humans and mice lends support to its potential role in atherosclerosis.


Assuntos
Aterosclerose/genética , Análise Mutacional de DNA , Ligação Genética , Mutação , Linhagem , Proteoglicanas/genética , Idoso , Animais , Apolipoproteínas E/deficiência , Aterosclerose/patologia , Espaço Extracelular/metabolismo , Feminino , Humanos , Masculino , Camundongos , Pessoa de Meia-Idade , Simulação de Dinâmica Molecular , Conformação Proteica , Proteoglicanas/química
18.
Circ Res ; 114(1): 193-204, 2014 Jan 03.
Artigo em Inglês | MEDLINE | ID: mdl-24385512

RESUMO

Cardiovascular disease (CVD) remains a major burden for morbidity and mortality in the general population, despite current efficacious low-density lipoprotein-cholesterol-lowering therapies. Consequently, novel therapies are required to reduce this residual risk. Prospective epidemiological studies have shown that high-density lipoprotein-cholesterol (HDL-C) levels are inversely correlated with cardiovascular disease risk, and this initiated the quest for HDL-C-increasing therapies. Consequently, several different targets in HDL metabolism have been identified. Initial studies addressing the effect of cholesteryl ester transfer protein inhibition on cardiovascular disease outcome have been discontinued for reasons of futility or increased mortality. As of yet, 2 cholesteryl ester transfer protein inhibitors are still in phase III studies. Other HDL-based interventions, such as apolipoprotein A1-based compounds, ABC-transporter upregulators, selective peroxisome proliferator-activated receptor modulators and lecithin-cholesterol acyltransferase-based therapy, hold great promise for the future. The aim of this review is to provide a comprehensive overview of HDL-targeted pharmaceutical strategies in humans, both in early development as well as in late stage clinical trials.


Assuntos
Anticolesterolemiantes/uso terapêutico , Doenças Cardiovasculares/tratamento farmacológico , Lipoproteínas HDL/metabolismo , Animais , Anticolesterolemiantes/farmacologia , Doenças Cardiovasculares/metabolismo , Humanos , Metabolismo dos Lipídeos/efeitos dos fármacos
19.
Curr Atheroscler Rep ; 15(6): 326, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23591671

RESUMO

Prospective epidemiological studies have consistently reported an inverse association between HDL cholesterol (HDL-C) levels and the risk of cardiovascular disease (CVD). However, large intervention trials on HDL-C-increasing drugs and recent Mendelian randomization studies have questioned a causal relationship between HDL-C and atherosclerosis. HDL-C levels have been shown to be highly heritable, and the combination of HDL-C-associated SNPs in recent large-scale genome-wide association studies (GWAS) only explains a small proportion of this heritability. As a large part of our current understanding of HDL metabolism comes from genetic studies, further insights in this research field may aid us in elucidating HDL functionality in relation to CVD risk. In this review we focus on the question of whether genetically defined HDL-C levels are associated with risk of atherosclerosis. We also discuss the latest insights for HDL-C-associated genes and recent GWAS data.


Assuntos
Aterosclerose/genética , HDL-Colesterol/genética , Aterosclerose/metabolismo , HDL-Colesterol/metabolismo , Predisposição Genética para Doença , Humanos , Polimorfismo de Nucleotídeo Único , Fatores de Risco
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...