Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 80
Filter
1.
J Clin Lipidol ; 18(1): e90-e96, 2024.
Article in English | MEDLINE | ID: mdl-38030518

ABSTRACT

BACKGROUND: Familial hypercholesterolemia (FH) is associated with lifelong elevated plasma concentrations of low-density lipoprotein cholesterol (LDL-C) and high risk of premature coronary heart disease (CHD). Clinical recommendations and treatments have emerged to facilitate the management of FH patients. Their impact on the burden of FH is however not well documented. OBJECTIVE: To compare the burden of FH between patients hospitalized for a CHD event 25 years apart in the French-Canadian founder population. METHODS: Lipid profiles, cardiovascular risk factors, treatments and FH status of 2,029 patients consecutively hospitalized for an acute CHD event between 2017 and 2022 (2022 Cohort) were compared to those of 2,506 patients with angiographically-confirmed CHD who were admitted between 1995 and 1998 (1998 Cohort). RESULTS: At the time of admission, 24.6 % of CHD patients had LDL-C levels >5.0 mmol/L in 1998 compared to 1.4 % in 2022, and FH was diagnosed in 9.6 % of patients in the 1998 cohort compared to 5.5 % in 2022 (p<0.001). FH patients hospitalized for a CHD event were older in 2022 than in 1998 (p <0.001). The prevalence of premature CHD requiring a hospitalization significantly decreased from 1998 to 2022 (64.3% vs. 44.1 %, p<0.001). At the moment of admission, 18.2 % of FH patients had LDL-C concentration <2.0 mmol/L in 2022 vs 0 % in 1998 (p <0.001). CONCLUSIONS: Over 25 years, FH patients tend to be older and contribute to a lower proportion of hospitalizations for CHD in the French-Canadian founder population. Despite significant improvement in diagnosis and treatment, FH management remains however sub-optimal.


Subject(s)
Coronary Artery Disease , Hyperlipoproteinemia Type II , North American People , Humans , Cholesterol, LDL , Canada/epidemiology , Hyperlipoproteinemia Type II/complications , Hyperlipoproteinemia Type II/epidemiology , Coronary Artery Disease/complications , Risk Factors
2.
Curr Atheroscler Rep ; 25(12): 1101-1111, 2023 Dec.
Article in English | MEDLINE | ID: mdl-38095804

ABSTRACT

PURPOSE OF REVIEW: The role of the inhibition of ANGPTL3 in severe or refractory hypercholesterolemia is well documented, less in severe hyperTG. This review focuses on the preclinical and clinical development of ApoC-III inhibitors and ANGPTL3, 4, and 3/8 complex inhibitors for the treatment of severe or refractory forms of hypertriglyceridemia to prevent cardiovascular disease or other morbidities. RECENT FINDINGS: APOC3 and ANGPTL3 became targets for drug development following the identification of naturally occurring loss of function variants in families with a favorable lipid profile and low cardiovascular risk. The inhibition of ANGPTL3 covers a broad spectrum of lipid disorders from severe hypercholesterolemia to severe hypertriglyceridemia, while the inhibition of ApoC-III can treat hypertriglyceridemia regardless of the severity. Preclinical and clinical data suggest that ApoC-III inhibitors, ANGPTL3 inhibitors, and inhibitors of the ANGPTL3/8 complex that is formed postprandially are highly effective for the treatment of severe or refractory hypertriglyceridemia. Inhibition of ANGPTL3 or the ANGPTL3/8 complex upregulates LPL and facilitates the hydrolysis and clearance of triglyceride-rich lipoproteins (TRL) (LPL-dependent mechanisms), whereas ApoC-III inhibitors contribute to the management and clearance of TRL through both LPL-dependent and LPL-independent mechanisms making it possible to successfully lower TG in subjects completely lacking LPL (familial chylomicronemia syndrome). Most of these agents are biologicals including monoclonal antibodies (mAb), antisense nucleotides (ASO), small interfering RNA (siRNA), or CRISPR-cas gene editing strategies.


Subject(s)
Hypercholesterolemia , Hyperlipidemias , Hypertriglyceridemia , Humans , Angiopoietin-Like Protein 3 , Apolipoprotein C-III/genetics , Hypertriglyceridemia/drug therapy , Hypertriglyceridemia/metabolism , Triglycerides/metabolism
3.
Orphanet J Rare Dis ; 18(1): 167, 2023 06 27.
Article in English | MEDLINE | ID: mdl-37370069

ABSTRACT

BACKGROUND: The familial chylomicronemia syndrome (FCS) is an ultra rare disease caused by lipoprotein lipase (LPL) deficiency associated with potentially lethal acute pancreatitis risk. Thrombocytopenia (platelet count < 150,000 × 109/L) has been reported in patients with FCS, treated or not with volanesorsen, a second generation APOC3 anti-sense oligonucleotide. Chylomicrons are the lipoproteins delivering fat after a meal and FCS thus has a post-prandial origin. Platelet count and function have not been studied post-prandially in FCS. OBJECTIVE: To evaluate post-prandial fluctuations in the platelet count (PLC) and functional defects of hemostasis in FCS. METHODS: PLC, functional defects in hemostasis and hematologic variables were measured up-to 5 h after a meal in 6 homozygotes for FCS causing gene variants (HoLPL), 6 heterozygotes for LPL loss-of-function variants (HeLPL) and 7 normolipidemic controls. RESULTS: Hourly post-prandial PLC was significantly lower in HoLPL than in controls (P < 0.009). Compared to the other groups, the PLC tended to decrease rapidly (in the first hour) post-meal in HoLPL (P = 0.03) and remained lower than baseline 5-h post-meal (P = 0.02) whereas it tended to slightly increase in normolipidemic controls (P = 0.02). Platelet function was not affected by the prandial status. In HoLPL, post-prandial fluctuations in the PLC positively correlated with the lymphocyte count (P = 0.005) and negatively with neutrophil/lymphocyte ratio (NLR). CONCLUSION: The PLC decreases post-prandially in FCS (HoLPL), is not associated with changes in functional defects of hemostasis and correlates with the NLR, a marker of acute pancreatitis severity.


Subject(s)
Hyperlipoproteinemia Type I , Pancreatitis , Humans , Hyperlipoproteinemia Type I/genetics , Platelet Count , Acute Disease , Pancreatitis/genetics , Hemostasis , Triglycerides
4.
Clin Biochem ; 114: 67-72, 2023 Apr.
Article in English | MEDLINE | ID: mdl-36780934

ABSTRACT

INTRODUCTION: Sustained chylomicronemia is a defect in post-prandial triglyceride management characterized by severe hypertriglyceridemia (triglyceride > 10 mmol/L) due to functional or genetic defects in lipoprotein lipase (LPL)-mediated triglyceride-rich lipoprotein lipolysis. Familial chylomicronemia syndrome (FCS) is a rare mendelian form of chylomicronemia caused by loss-of-function variants in LPL or LPL-related genes. Most individuals with chylomicronemia however present multifactorial chylomicronemia (MCS), in which LPL bio-availability and activity are variable. FCS and MCS differ in terms of clinical characteristics and risk of disease, and diagnosis scoring systems have been proposed to accurately distinguish FCS from MCS. OBJECTIVE: The aim of this study was to assess the strength of the relationship between plasma post-heparin LPL activity and two published chylomicronemia diagnosis scoring systems. DESIGN AND METHODS: Post-heparin plasma LPL activity was measured using colorimetric assays in a sample of 29 subjects with sustained chylomicronemia (20 FCS and 9 MCS). Chylomicronemia diagnosis scores were obtained for all subjects using the scoring system A (model A), which integrates apolipoprotein B and free glycerol, a surrogate marker of triglyceride hydrolysis, and the scoring system B (model B). Correlation analyses were conducted to estimate the linear relationship between LPL activity and the two diagnosis scoring systems. RESULTS: There was a significant (p < 0.001) difference in post-heparin LPL activity between FCS and MCS. Both scoring systems significantly correlated with post-heparin LPL activity (model A: rs = -0.64, p < 0.001; model B: rs = -0.54, p = 0.002). CONCLUSIONS: These result suggest that chylomicronemia diagnosis scoring systems correlate with LPL activity and adequately contribute to distinguish FCS from MCS.


Subject(s)
Lipoprotein Lipase , Lipoproteins , Humans , Lipoprotein Lipase/genetics , Triglycerides , Heparin
5.
Can J Cardiol ; 39(5): 668-677, 2023 05.
Article in English | MEDLINE | ID: mdl-36586484

ABSTRACT

BACKGROUND: Limitations of the Friedewald equation for low-density-lipoprotein cholesterol (F-LDLC) calculation led to the Martin-Hopkins (M-LDLC) and Sampson-National Institutes of Health (S-LDLC) equations. We studied these newer calculations of LDLC for correlation and discordance for stratification into the Canadian Cardiovascular Society (CCS) 2021 Dyslipidemia Guidelines' cardiovascular disease (CVD) risk categories. METHODS: We performed analyses on lipid profiles from 3 populations: records of a hospital biochemistry laboratory (population 1), lipid clinic patients without select monogenic dyslipidemias (population 2A), and lipid clinic patients with familial hypercholesterolemia (FH; population 2B). RESULTS: There was very strong correlation among the 3 calculated LDLC. In populations 1 and 2A, M-LDLC and S-LDLC were progressively higher than F-LDLC as triglyceride (TG) levels increased from normal to ∼ 5 mmol/L. In population 2B, M-LDLC was higher than F-LDLC, but S-LDLC was progressively lower than F-LDLC. Using the CCS 2021 guidelines' 4 CVD risk categories, 7.0% (population 2A) to 7.2% (population 1) of cases for M-LDLC vs F-LDLC and 3.9% (population 2A) to 4.4% (population 1) of cases for S-LDLC vs F-LDLC were reclassified to an adjacent CVD risk category, mostly from a lower to a higher risk category. CONCLUSIONS: Switching from F-LDLC to S-LDLC or M-LDLC can reclassify up to ∼ 4.4% or 7.2% of patients, respectively, to another CCS CVD risk category. The difference between F-LDLC and M-LDLC or S-LDLC is greater with higher TG, and with lower LDLC. We recommend that clinical laboratories switch to reporting results from either M-LDLC or S-LDLC, but S-LDLC should not be used in FH patients, pending further studies.


Subject(s)
Cardiovascular Diseases , Cholesterol, LDL , Dyslipidemias , Hyperlipoproteinemia Type II , Humans , Canada/epidemiology , Cardiovascular Diseases/epidemiology , Cardiovascular Diseases/etiology , Dyslipidemias/epidemiology , Triglycerides
6.
J Endocr Soc ; 6(8): bvac103, 2022 Aug 01.
Article in English | MEDLINE | ID: mdl-35860753

ABSTRACT

Context: Palmar striated xanthomas (PSX) are macular subcutaneous lesions conferring a yellow-to-orange coloration of palmar and finger creases that characterize dysbetalipoproteinemia, a disease associated with sustained plasma accumulation of triglyceride-rich lipoprotein remnants. Although remnants accumulation may occur in any condition interfering with triglyceride-rich lipoprotein hydrolysis or clearance, the presence of PSX has not been systematically assessed across the spectrum of lipid disorders potentially associated with sustained or recurrent remnants accumulation. Objective: The aim of this study was to assess the occurrence of (PSX) in a wide spectrum of lipid disorders ranging from very severe hypercholesterolemia (homozygous familial hypercholesterolemia) to very severe hypertriglyceridemia (chylomicronemia). Methods: This study involved 3382 dyslipidemic White adult patients (1856 men and 1526 women) seen at the Chicoutimi Hospital Lipid Clinic (Quebec, Canada), covering a wide range of lipid disorders, from severe hypertriglyceridemia to severe hypercholesterolemia. Categorical variables were compared using the Pearson χ 2 statistic, whereas univariate analysis of variance or nonparametric Kruskal-Wallis was used for continuous variables. Results: A total of 5.1% (173/3382) of the studied patients presented PSX, a majority of them (67.1%) being women. PSX were observed in 18.8% of patients with dysbetalipoproteinemia and also among 14.1% of hypertriglyceridemic patients with partial lipoprotein lipase deficiency, 3.7% of patients with chylomicronemia, and in all those with homozygous familial hypercholesterolemia. Overall, 10.7% of patients with PSX did not meet dysbetalipoproteinemia diagnosis criteria. Conclusion: According to our study, the PSX prevalence estimate among patients without dysbetalipoproteinemia would be around 10% and they could be observed in a wide spectrum of lipid disorders associated with recurrent or sustained remnant lipoprotein accumulation.

7.
Can J Diabetes ; 46(5): 480-486, 2022 Jul.
Article in English | MEDLINE | ID: mdl-35718690

ABSTRACT

OBJECTIVES: The impact of type 2 diabetes (T2DM) on biomarkers denoting lipoprotein compositional status was studied in mild and moderate hypertriglyceridemia (HTG). Diabetic dyslipidemia pathophysiology could contribute to differences in lipoprotein compositional status, which could be reflected in the preferred cardiovascular disease risk prediction markers in HTG: non-high-density lipoprotein cholesterol (non-HDLC) and apolipoprotein B (apoB). METHODS: A total of 2,775 fasting lipid profiles from a tertiary care lipid clinic were analyzed as 2 subgroups (with and without T2DM), stratified by triglyceride (TG) levels: normotriglyceridemia (TG 0.01 to 1.7 mmol/L), mild HTG (TG 1.71 to 5 mmol/L) and moderate HTG (TG 5.01 to 10 mmol/L). The mean non-HDLC:apoB ratio in each TG stratum and subgroup was analyzed. We also used linear regression to assess the correlation between non-HDLC and apoB. RESULTS: The mean non-HDLC:apoB ratio was increased in both subgroups in patients with mild and moderate HTG, compared to those with normotriglyceridemia. In moderate HTG, the mean non-HDLC:apoB ratio in the subgroup with T2DM was significantly lower than the subgroup without T2DM. In mild and moderate HTG, the subgroup with T2DM had a stronger correlation between non-HDLC and apoB than did the subgroup without T2DM. DISCUSSION AND CONCLUSIONS: In mild and moderate HTG, adults with T2DM exhibit lipid profiles that represent a different and more atherogenic lipoprotein compositional status, when compared with adults without T2DM. For the same severity of HTG, the lipoprotein compositional status in diabetic dyslipidemia suggests that there is increased abundance of smaller non-HDL particles and their remnants, which are highly atherogenic.


Subject(s)
Diabetes Mellitus, Type 2 , Dyslipidemias , Hyperlipidemias , Hypertriglyceridemia , Adult , Apolipoproteins B , Cholesterol , Diabetes Mellitus, Type 2/complications , Diabetes Mellitus, Type 2/epidemiology , Humans , Hypertriglyceridemia/complications , Lipoproteins , Triglycerides
8.
Can J Cardiol ; 38(3): 311-319, 2022 03.
Article in English | MEDLINE | ID: mdl-34774719

ABSTRACT

BACKGROUND: Whether low-density lipoprotein (LDL) receptor (LDLR) residual activity influences the LDL-lowering effect of statins in heterozygous familial hypercholesterolemia (HeFH) remains unclear. The objective of this study was to investigate the relationship between the LDLR genotype and statin-induced LDL cholesterol (LDL-C) reductions in HeFH. METHODS: A total of 615 individuals with HeFH (receptor-defective [RD] genotype: n = 226; receptor-negative [RN] genotype: n = 389) from 7 lipid clinics across Canada who initiated statin monotherapy were included in this retrospective longitudinal study. Statin-induced reductions in LDL-C among individuals with RD and RN genotypes were compared with the use of linear models. RESULTS: There were 334 women and 281 men with a mean untreated LDL-C concentrations of 6.97 ± 1.65 mmol/L. Untreated and on-statin LDL-C levels where higher among patients with an RN genotype: untreated: RN 7.24 (95% confidence interval [CI] 6.98-7.50) mmol/L vs RD 6.70 (95% CI 6.41-6.98) mmol/L (P = 0.0002); on-statin: RN 4.50 (95% CI 4.31-4.70) vs RD 4.05 (95% CI 3.84-4.26) mmol/L (P = 0.0004). After adjustments for age, sex, smoking status, untreated LDL-C concentrations, statin type and dose, as well as the clinic where the patients were treated, the LDL-C-lowering effect of statins was significantly weaker for individuals with an RN mutation than for individuals with an RD mutation: RN: -31.1% (95% CI -34.7% to -27.4) vs RD -36.5% (95% CI -40.4% to -32.6%); P < 0.0001. The LDLR genotype was the strongest nonmodifiable independent correlate of statin-induced LDL-C reductions (R2 = 2.3%; P = 0.0001). CONCLUSION: The LDLR genotype is significantly associated with statin-induced reductions in LDL-C concentrations in HeFH.


Subject(s)
Cholesterol, LDL/blood , Hydroxymethylglutaryl-CoA Reductase Inhibitors/administration & dosage , Hyperlipoproteinemia Type II , Lipid Metabolism , Receptors, LDL/genetics , Canada/epidemiology , Female , Genetic Profile , Heterozygote , Humans , Hyperlipoproteinemia Type II/blood , Hyperlipoproteinemia Type II/drug therapy , Hyperlipoproteinemia Type II/epidemiology , Hyperlipoproteinemia Type II/genetics , Lipid Metabolism/drug effects , Lipid Metabolism/genetics , Longitudinal Studies , Male , Middle Aged , Mutation , Pharmacogenomic Testing
9.
J Med Genet ; 58(10): 653-665, 2021 10.
Article in English | MEDLINE | ID: mdl-33910931

ABSTRACT

The Saguenay-Lac-Saint-Jean (SLSJ) region located in the province of Quebec was settled in the 19th century by pioneers issued from successive migration waves starting in France in the 17th century and continuing within Quebec until the beginning of the 20th century. The genetic structure of the SLSJ population is considered to be the product of a triple founder effect and is characterised by a higher prevalence of some rare genetic diseases. Several studies were performed to elucidate the historical, demographic and genetic background of current SLSJ inhabitants to assess the origins of these rare disorders and their distribution in the population. Thanks to the development of new sequencing technologies, the genes and the variants responsible for the most prevalent conditions were identified. Combined with other resources such as the BALSAC population database, identifying the causal genes and the pathogenic variants allowed to assess the impacts of some of these founder mutations on the population health and to design precision medicine public health strategies based on carrier testing. Furthermore, it stimulated the establishment of many public programmes.We report here a review and an update of a subset of inherited disorders and founder mutations in the SLSJ region. Data were collected from published scientific sources. This work expands the knowledge about the current frequencies of these rare disorders, the frequencies of other rare genetic diseases in this population, the relevance of the carrier tests offered to the population, as well as the current available treatments and research about future therapeutic avenues for these inherited disorders.


Subject(s)
Founder Effect , Genetic Diseases, Inborn/epidemiology , Genetic Diseases, Inborn/genetics , Genetic Predisposition to Disease , France , Genes, Recessive , Genetic Background , Genetic Diseases, Inborn/diagnosis , Genetic Testing , Humans , Mass Screening , Phenotype , Prevalence , Quebec/epidemiology
10.
J Clin Med ; 10(4)2021 Feb 09.
Article in English | MEDLINE | ID: mdl-33572376

ABSTRACT

Non-alcoholic fatty liver disease (NAFLD) is frequent in patients with features of the metabolic syndrome (MetS), obesity, or type 2 diabetes. Lipoprotein lipase (LPL) is the main driver of triglyceride (TG) hydrolysis in chylomicrons and very-low density lipoproteins (VLDL). In some patients with MetS, dysfunction of this pathway can lead to plasma TG values > 10 mmol/L (multifactorial chylomicronemia or MCS). Chylomicronemia also characterizes LPL deficiency (LPLD), a rare autosomal recessive disease called familial chylomicronemia syndrome (FCS), which is associated with an increased risk of recurrent pancreatitis. This study aims to investigate the expression of NAFLD, as assessed by transient elastography, in MCS and FCS subjects. Data were obtained from 38 subjects with chylomicronemia; 19 genetically confirmed FCS and 19 sex- and age-matched MCS. All participants underwent liver ultrasonography and stiffness measurement after a 4-h fast using transient elastography (FibroScan®, Echosens, Waltham, MA, USA). NAFLD (controlled attenuation parameter (CAP) > 280 dB/m) was observed in 42.1% of FCS and 73.7% of MCS subjects (p = 0.05). FCS subjects had lower body mass index (BMI) than MCS. Only 25% of FCS subjects with NAFLD had a BMI ≥ 30 compared to 64.3% in MCS (p = 0.004). In FCS, NAFLD occurred even in the presence of very low (≤18 kg/m2) BMI. In both FCS and MCS, CAP was negatively associated with acute pancreatitis risk. In this study, NAFLD was commonly observed in both FCS and MCS subjects and occurred independently of the BMI and fasting glucose values in FCS; NAFLD was associated with a lower occurrence of acute pancreatitis episodes.

11.
J Clin Med ; 10(1)2020 Dec 27.
Article in English | MEDLINE | ID: mdl-33375401

ABSTRACT

Familial hypercholesterolemia (FH) is an autosomal dominant trait characterized by elevated low-density lipoprotein-cholesterol (LDL-C) concentrations appearing at birth and is associated with increased risk of premature atherosclerotic cardiovascular disease (CVD). However, in some cases, FH subjects over 70 years of age have surprisingly never experienced any CVD symptoms throughout their entire lives. The objective of this study consists of identifying biological and environmental markers acting as cardioprotective factors and associated with unexpected survival in FH. Upon age and reported cardiovascular events (CVE) stratification, we identified a total of 458 French-Canadian FH subjects with premature reported CVE, and 1297 young adults as well as 24 elderly subjects (≥70 years) who have never reported CVE requiring hospitalization. Logistic regression models were used to depict cardioprotective markers among FH survivors (≥70 years). Regression analyses of the FH cohort showed that female sex (odds ratio (OR) = 12.92 (4.23-39.46); p < 0.0001), high levels of high-density lipoprotein (HDL)-C (OR = 6.76 (2.43-18.79); p = 0.0002) and elevated concentrations of adiponectin (OR = 71.40 (5.20-980.47); p = 0.001) were significant contributory factors in reducing FH-related CVD risk. Notably, female (OR = 11.45 (1.25-105.98); p = 0.031) and high HDL-C (OR = 9.78 (1.75-54.67); p = 0.009) were shown to be significant covariates associated with survival in FH. Non-smoking (OR = 11.73 (4.36-31.56); p < 0.0001) was also identified as an environmental factor associated with CVE-free survival. Based on this configured model of premature CVE occurrence, these results demonstrated that, beyond LDL-C levels, female sex, high HDL-C, elevated adiponectin and non-smoking are important markers that contribute to a reduced risk of CVD and CVE-free survival in FH.

12.
J Clin Lipidol ; 14(6): 825-836, 2020.
Article in English | MEDLINE | ID: mdl-33032940

ABSTRACT

BACKGROUND: Non-high density lipoprotein cholesterol (non-HDLC) represents the cholesterol in triglyceride-rich lipoproteins (TRL) and low-density lipoproteins (LDL). Apolipoprotein B (apoB) reflects the number of TRL and LDL particles. In hypertriglyceridemia (HTG), there is triglyceride (TG) enrichment of TRLs, and also a substantial increase of cholesterol in larger TRLs that considerably augments the non-HDLC value. Therefore, in HTG, non-HDLC could increase disproportionately with respect to apoB. OBJECTIVE: We aimed to compare the relative effect of the full range of mild, moderate, and severe HTG on the status of non-HDLC and apoB as cardiovascular disease (CVD) risk markers. METHODS: Analysis of lipid profile data from 4347 patients in a Lipid Clinic cohort with baseline fasting lipid profiles documented prior to starting lipid-lowering medications. The correlation between non-HDLC and apoB was assessed in intervals of increasing TG. Non-HDLC and apoB were analyzed at each TG level using comparative CVD risk equivalent categories and assessed for divergence and discordance. RESULTS: With increasing TG levels: (1) the correlation between non-HDLC and apoB diminished progressively, (2) non-HDLC levels increased continuously, whereas apoB levels plateaued after an initial increase up to TG of ~ 4.0-5.0 mmol/L (~354-443 mg/dL), (3) there was divergence in the stratification of non-HDLC and apoB into CVD risk equivalent categories. CONCLUSIONS: Non-HDLC and apoB should not be viewed as interchangeable CVD risk markers in the presence of severe HTG. This has never been tested. With increasing HTG severity, discordance between non-HDLC and apoB can cause clinically important divergence in CVD risk categorization.


Subject(s)
Apolipoproteins B/metabolism , Cardiovascular Diseases/complications , Cholesterol/metabolism , Hypertriglyceridemia/complications , Hypertriglyceridemia/metabolism , Adult , Biomarkers/metabolism , Female , Humans , Male , Middle Aged , Risk Factors
13.
Atherosclerosis ; 310: 54-63, 2020 10.
Article in English | MEDLINE | ID: mdl-32906018

ABSTRACT

BACKGROUND AND AIMS: Homozygous familial hypercholesterolemia (HoFH) is an orphan disease, most often caused by bi-allelic mutations of the LDLR gene. Patients with HoFH have elevated LDL-C levels >13 mmol/L, tendinous xanthomata and severe, premature atherosclerotic cardiovascular disease (ASCVD). Untreated, most HoFH patients die of ASCVD in youth. New therapeutic modalities include lomitapide, an inhibitor of microsomal triglyceride transfer protein that lowers hepatic LDL-C production. We have recently identified 79 Canadian patients with HoFH. Here, we describe our experience with lomitapide in the province of Quebec, a geographic area known to have a high prevalence of HoFH. METHODS: This is a retrospective case series of 12 HoFH patients followed at three lipidology centers in the province of Quebec. RESULTS: Mean age of the patients was 44 ± 18 years; age at time of HoFH diagnosis ranged from 2 to 59 years. All patients were on a statin and ezetimibe 10 mg/day and five patients were treated with LDL apheresis. Treatment with lomitapide reduced LDL-C levels by 38% (intention-to-treat). Intolerable gastrointestinal side effects were observed in 3/12 patients and were the main reason for treatment discontinuation. Three patients tolerated lomitapide at doses ranging between 5 and 30 mg/day without major side effects. Downwards drug titration was necessary in the 6 remaining patients because of gastrointestinal side effects (n = 5) and elevated liver enzymes (n = 1), and 2 of them finally discontinued treatment. CONCLUSIONS: Lomitapide may be used to further decrease LDL-C in HoFH patients; gastrointestinal side effects and hepatic toxicity may limit adherence.


Subject(s)
Anticholesteremic Agents , Hyperlipoproteinemia Type II , Adult , Anticholesteremic Agents/adverse effects , Benzimidazoles , Canada , Homozygote , Humans , Hyperlipoproteinemia Type II/diagnosis , Hyperlipoproteinemia Type II/drug therapy , Hyperlipoproteinemia Type II/genetics , Middle Aged , Quebec , Retrospective Studies
14.
Endocr J ; 67(11): 1157-1161, 2020 Nov 28.
Article in English | MEDLINE | ID: mdl-32727960

ABSTRACT

A fasting triglyceridemia >10 mmol/L is associated with chylomicronemia (CM) and an increased recurrent acute pancreatitis (RAP) risk. The number of pancreatitis episodes varies significantly between patients with CM. The objective of this study was to investigate gene expression profiles of RAP in patients with CM. A total of 47 CM subjects participated in this study. Prior to the analyses, all patients were divided into three groups covering a wide spectrum of RAP: 0 (n = 21), 1-3 (n = 10) or >4 (n = 16) pancreatitis episodes. Gene expression profiles were compared to those of 15 healthy normolipidemic controls. Differential expression moderated T-tests between studied groups were performed using a linear model of the Bioconductor package Limma. The False discovery rate was controlled using the Benjamini-Hochberg procedure. At a p-value <0.01, a false discovery rate of 5% and a >2-fold change expression significance levels, a set of 41 probes have been found differentially expressed in CM subjects with no pancreatitis, 103 in the CM group with 1 to 3 pancreatitis, and 94 in the group with ≥4 pancreatitis compared to healthy controls. Of the identified annotated probes, 14 are shared by all CM groups; 3 are specific to CM with no pancreatitis; 11 are specific to CM with 1 to 3 pancreatitis, and 17 are specific to CM with ≥4 pancreatitis. Most of the annotated biomarkers are involved in inflammatory, immune, lipoprotein kinetics or signalling biological pathways. These results reveal gene expression signatures of RAP in patients with CM.


Subject(s)
Hyperlipoproteinemia Type I/genetics , Hypertriglyceridemia/genetics , Pancreatitis/genetics , Transcriptome , Adult , Aged , Case-Control Studies , Chylomicrons/blood , Female , Gene Expression Profiling , Humans , Male , Middle Aged , Recurrence
15.
J Endocr Soc ; 4(6): bvaa056, 2020 Jun 01.
Article in English | MEDLINE | ID: mdl-32537545

ABSTRACT

Familial chylomicronemia syndrome (FCS) is a rare disorder associated with chylomicronemia (CM) and an increased risk of pancreatitis. Most individuals with CM do not have FCS but exhibit multifactorial CM (MCM), which differs from FCS in terms of risk and disease management. This study aimed to investigate clinical and gene expression profiles of FCS and MCM patients. Anthropometrics, clinical, and biochemical variables were analyzed in 57 FCS and 353 MCM patients. Gene expression analyses were performed in a subsample of 19 FCS, 28 MCM, and 15 normolipidemic controls. Receiver operating characteristic (ROC) curve analyses were performed to analyze the capacity of variables to discriminate FCS from MCM. Sustained fasting triglycerides ≥20 mmol/L (>15 mmol/L with eruptive xanthomas), history of pancreatitis, poor response to fibrates, diagnosis of CM at childhood, body mass index <22 kg/m2, and delipidated apolipoprotein B or glycerol levels <0.9 g/L and <0.05 mmol/L, respectively, had an area under the ROC curve ≥0.7. Gene expression analyses identified 142 probes differentially expressed in FCS and 32 in MCM compared with controls. Among them, 13 probes are shared between FCS and MCM; 63 are specific to FCS and 2 to MCM. Most FCS-specific or shared biomarkers are involved in inflammatory, immune, circadian, postprandial metabolism, signaling, docking systems, or receptor-mediated clearance mechanisms. This study reveals differential signatures of FCS and MCM. It opens the door to the identification of key mechanisms of CM expression and potential targets for the development of new treatments.

16.
Lipids Health Dis ; 19(1): 98, 2020 May 19.
Article in English | MEDLINE | ID: mdl-32430061

ABSTRACT

BACKGROUND: Increased apolipoprotein (apo) B level (hyperapoB) is a strong predictor of cardiovascular disease (CVD), even in patients who achieve recommended LDL-Cholesterol (LDL-C) goals. ApoB level, an important correlate of metabolic syndrome (MetS), is influenced by several gene-environment interactions. Some of them are rare and can explain a large proportion of apoB variance, whereas others more common have variable effects. The aim of this study was to evaluate the association of interaction between smoking and common hyperapoB gene variants (PPARα-L162V, lipoprotein lipase loss-of function mutation, apo e4 allele or apo E2/2 genotype) with plasma apoB concentrations, according to the expression of MetS. METHODS: This study was performed among 1798 subjects. Smoking was defined as non/mild smokers vs. moderate-to-heavy smokers. ApoB levels were determined using nephelometry. Logistic regression models were used to document interactions between smoking habits and the presence of hyperapoB gene variants on the relative odds to exhibit increased plasma apoB concentrations. RESULTS: Around 29% of individuals with a low-risk lipid profile without MetS component had hyperapoB. Smoking and the presence of hyperapoB gene variants tended to be associated with higher plasma apoB levels even in presence of low-LDL-C. There was a significant interaction (P = 0.04) between the presence of ≥1 gene variants and smoking on the risk to exhibit hyperapoB among subjects with low risk profile in primary prevention. CONCLUSIONS: Combination of life habits assessment and some common genes variants may detect a significant proportion of patients with increased apoB levels, and therefore a higher risk of CVD, who could have been initially perceived as low-risk.


Subject(s)
Apolipoprotein E4/genetics , Apolipoproteins B/blood , Gene-Environment Interaction , Lipoprotein Lipase/genetics , PPAR alpha/genetics , Polymorphism, Genetic , Smoking/genetics , Female , Humans , Male , Metabolic Syndrome , Middle Aged , Quebec
17.
J Endocr Soc ; 4(1): bvz010, 2020 Jan 01.
Article in English | MEDLINE | ID: mdl-32010872

ABSTRACT

CONTEXT: Calculated non-high-density lipoprotein (HDL) cholesterol (non-HDLC) should selectively include cholesterol from atherogenic lipoproteins to be a reliable risk marker of cardiovascular disease. In hypertriglyceridemia (HTG), there is increased abundance of larger and less atherogenic triglyceride-rich lipoproteins (TRL), namely, larger very-low-density lipoproteins (VLDL), and chylomicrons. OBJECTIVE: We aim to demonstrate that serum triglyceride (TG) level has a substantial impact on non-HDLC's ability to represent cholesterol from atherogenic lipoproteins, even though TG is not part of the calculation for non-HDLC. DESIGN: Analysis of lipid profile data. SETTINGS: Lipid Clinic patient cohort, and Biochemistry Laboratory patient cohort. PATIENTS OR OTHER PARTICIPANTS: 7,492 patients in the Lipid Clinic cohort with baseline lipid profiles documented prior to starting lipid-lowering medications and 156,311 lipid profiles from The Ottawa Hospital Biochemistry Laboratory cohort. INTERVENTION: None. MAIN OUTCOME MEASURE: Our modeling process includes derivation of TG-interval-specific lipoprotein composition factor (LCF) for TRL, which represents the mass ratio of cholesterol to TG in TRL. A high LCF indicates that the TRLs are mainly the cholesterol-rich atherogenic remnant lipoproteins. A low LCF indicates that the TRLs are mainly the TG-rich larger VLDL and chylomicrons. RESULTS: As serum TG increases, there is progressive decline in the LCF for TRL, which indicates that the calculated non-HDLC level reflects progressive inclusion of cholesterol from larger TRL. This is shown in both cohorts. CONCLUSIONS: Calculated non-HDLC is influenced by TG level. As TG increases, non-HDLC gradually includes more cholesterol from larger TRL, which are less atherogenic than LDL and remnant lipoproteins.

18.
Expert Opin Drug Discov ; 15(4): 403-414, 2020 04.
Article in English | MEDLINE | ID: mdl-31973581

ABSTRACT

Introduction: Proprotein convertase subtilisin/kexin type 9 (PCSK9) is a serine protease that binds and promotes the lysosomal degradation of the low-density lipoprotein receptors (LDLR). Upon its discovery in 2002, PCSK9 inhibition has subsequently emerged as a novel target for lowering LDL-cholesterol (LDL-C) and reducing coronary heart disease. Evolocumab, a monoclonal antibody directed against human PCSK9, was approved in 2015 as an adjunct to lipid-lowering therapy for treating patients with familial hypercholesterolemia (FH) or patients with high cardiovascular risk, who are treated with maximally tolerated lipid-lowering agents and have not reached the recommended LDL-C levels.Areas covered: The authors illustrate the rapid pace of the drug development process that monoclonal antibodies, including evolocumab, have demonstrated during the last decade. In less than 15 years from its discovery, this lipid-lowering target has successfully progressed from bench-side to clinical practice and has been recently approved to reduce cardiovascular events in patients with established atherosclerotic cardiovascular disease (ASCVD).Expert opinion: Evolocumab has demonstrated a good safety profile and robust efficacy in terms of its lipid-lowering effect and ASCVD risk reduction, yet affordability, accessibility, and cost-effectiveness of PCSK9 monoclonal antibodies remain a hurdle in the 'real-world' setting. These challenges facing the upcoming generation of precision medicine therapies must be addressed upfront.


Subject(s)
Antibodies, Monoclonal, Humanized/administration & dosage , Anticholesteremic Agents/administration & dosage , Hypercholesterolemia/drug therapy , Animals , Antibodies, Monoclonal, Humanized/adverse effects , Antibodies, Monoclonal, Humanized/pharmacology , Anticholesteremic Agents/adverse effects , Anticholesteremic Agents/pharmacology , Atherosclerosis/prevention & control , Cholesterol, LDL/blood , Cost-Benefit Analysis , Drug Development , Humans , PCSK9 Inhibitors
19.
J Dev Orig Health Dis ; 11(4): 369-378, 2020 08.
Article in English | MEDLINE | ID: mdl-31753053

ABSTRACT

Placental lipids transfer is essential for optimal fetal development, and alterations of these mechanisms could lead to a higher risk of adverse birth outcomes. Low-density lipoprotein receptor (LDLR), LDL receptor-related protein 1 (LRP1), and scavenger receptor class B type 1 (SCARB1) genes are encoding lipoprotein receptors expressed in the placenta where they participate in cholesterol exchange from maternal to fetal circulation. The aim of this study was thus to investigate the association between maternal lipid changes occurring in pregnancy, placental DNA methylation (DNAm) variations at LDLR, LRP1, and SCARB1 gene loci, and newborn's anthropometric profile at birth. Sixty-nine normoglycemic women were followed from the first trimester of pregnancy until delivery. Placental DNAm was quantified at 43 Cytosine-phosphate-Guanines (CpGs) at LDLR, LRP1, and SCARB1 gene loci using pyrosequencing: 4 CpGs were retained for further analysis. Maternal clinical data were collected at each trimester of pregnancy. Newborns' data were collected from medical records. Statistical models included minimally newborn sex and gestational and maternal age. Maternal total cholesterol changes during pregnancy (ΔT3-T1) were correlated with DNAm variations at LDLR (r = -0.32, p = 0.01) and LRP1 (r = 0.34, p = 0.007). DNAm at these loci was also correlated with newborns' cord blood triglyceride and leptin levels. Mediation analysis supports a causal relationship between maternal cholesterol changes, DNAm levels at LRP1 locus, and cord blood leptin concentration (pmediation = 0.02). These results suggest that LRP1 DNAm link maternal blood cholesterol changes in pregnancy and offspring adiposity at birth, which provide support for a better follow-up of blood lipids in pregnancy.


Subject(s)
Cholesterol/metabolism , DNA Methylation , Epigenesis, Genetic , Fetal Blood/metabolism , Leptin/metabolism , Low Density Lipoprotein Receptor-Related Protein-1/genetics , Placenta/metabolism , Adult , Female , Gene Expression Regulation , Humans , Infant, Newborn , Male , Pregnancy , Pregnancy Trimesters , Prognosis , Receptors, LDL/genetics , Scavenger Receptors, Class B/genetics
20.
PLoS One ; 14(6): e0218115, 2019.
Article in English | MEDLINE | ID: mdl-31242253

ABSTRACT

AIMS: Statin-related myopathy (SRM), which includes rhabdomyolysis, is an uncommon but important adverse drug reaction because the number of people prescribed statins world-wide is large. Previous association studies of common genetic variants have had limited success in identifying a genetic basis for this adverse drug reaction. We conducted a multi-site whole-exome sequencing study to investigate whether rare coding variants confer an increased risk of SRM. METHODS AND RESULTS: SRM 3-5 cases (N = 505) and statin treatment-tolerant controls (N = 2047) were recruited from multiple sites in North America and Europe. SRM 3-5 was defined as symptoms consistent with muscle injury and an elevated creatine phosphokinase level >4 times upper limit of normal without another likely cause of muscle injury. Whole-exome sequencing and variant calling was coordinated from two analysis centres, and results of single-variant and gene-based burden tests were meta-analysed. No genome-wide significant associations were identified. Given the large number of cases, we had 80% power to identify a variant with minor allele frequency of 0.01 that increases the risk of SRM 6-fold at genome-wide significance. CONCLUSIONS: In this large whole-exome sequencing study of severe statin-related muscle injury conducted to date, we did not find evidence that rare coding variants are responsible for this adverse drug reaction. Larger sample sizes would be required to identify rare variants with small effects, but it is unclear whether such findings would be clinically actionable.


Subject(s)
Hydroxymethylglutaryl-CoA Reductase Inhibitors/adverse effects , Muscle, Skeletal , Rhabdomyolysis , Whole Genome Sequencing , Genome-Wide Association Study , High-Throughput Nucleotide Sequencing , Humans , Hydroxymethylglutaryl-CoA Reductase Inhibitors/therapeutic use , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Rhabdomyolysis/chemically induced , Rhabdomyolysis/genetics , Rhabdomyolysis/metabolism , Rhabdomyolysis/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...