1
|
Boffa MB, Koschinsky ML. Lipoprotein(a) and cardiovascular disease. Biochem J 2024; 481:1277-1296. [PMID: 39302109 PMCID: PMC11555715 DOI: 10.1042/bcj20240037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Revised: 08/28/2024] [Accepted: 08/28/2024] [Indexed: 09/22/2024]
Abstract
Elevated plasma levels of lipoprotein(a) (Lp(a)) are a prevalent, independent, and causal risk factor for atherosclerotic cardiovascular disease and calcific aortic valve disease. Lp(a) consists of a lipoprotein particle resembling low density lipoprotein and the covalently-attached glycoprotein apolipoprotein(a) (apo(a)). Novel therapeutics that specifically and potently lower Lp(a) levels are currently in advanced stages of clinical development, including in large, phase 3 cardiovascular outcomes trials. However, fundamental unanswered questions remain concerning some key aspects of Lp(a) biosynthesis and catabolism as well as the true pathogenic mechanisms of the particle. In this review, we describe the salient biochemical features of Lp(a) and apo(a) and how they underlie the disease-causing potential of Lp(a), the factors that determine plasma Lp(a) concentrations, and the mechanism of action of Lp(a)-lowering drugs.
Collapse
Affiliation(s)
- Michael B. Boffa
- Department of Biochemistry, University of Western Ontario, London, Ontario, Canada
- Robarts Research Institute, University of Western Ontario, London, Ontario, Canada
| | - Marlys L. Koschinsky
- Robarts Research Institute, University of Western Ontario, London, Ontario, Canada
- Department of Physiology and Pharmacology, University of Western Ontario, London, Ontario, Canada
| |
Collapse
|
2
|
Krishnamurthy HK, Rajavelu I, Reddy S, Pereira M, Jayaraman V, Krishna K, Song Q, Wang T, Bei K, Rajasekaran JJ. Association of Apolipoprotein E (APOE) Polymorphisms With Serological Lipid and Inflammatory Markers. Cureus 2024; 16:e60721. [PMID: 38903305 PMCID: PMC11187349 DOI: 10.7759/cureus.60721] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/13/2024] [Indexed: 06/22/2024] Open
Abstract
Background The study aims to assess the association of apolipoprotein E (APOE) gene polymorphisms with serological lipid and inflammatory markers to determine their potential role in predicting the risk of cardiovascular diseases (CVDs) and Alzheimer's disease (AD). Methodology A total of 915 individuals underwent testing for lipid and inflammatory biomarkers at Vibrant America Clinical Laboratory. Clinical data, blood lipid and inflammatory profiles, and APOE genotyping were analyzed using polymerase chain reaction-restriction fragment length polymorphism (PCR-RFLP). Results Compared to the E3/E3 genotype, individuals with E2/E3 genotypes showed higher levels of high-density lipoprotein (HDL), triglycerides, apolipoprotein A (APOA), high-sensitivity C-reactive protein (hs-CRP), and myeloperoxidase (MPO). E2/E4 genotype carriers had higher levels of HDL, triglycerides, Lp(a), and N-terminal pro b-type natriuretic peptide (BNPNT). E3/E4 genotypes were associated with elevated levels of total cholesterol, LDL, Lp(a), hs-CRP, small-density low-density lipoprotein (SDLDL), oxidized LDL (OXLDL), MPO, LDL-CAL, PLAC, and APOB. The E4/E4 group displayed higher concentrations of total cholesterol, LDL, APOB, Lp(a), hs-CRP, SDLDL, OXLDL, MPO, LDLCAL, and PLAC compared to E3/E3 carriers. These findings highlight the potential atherogenic effect of the ε4 allele and the protective effect of the ε2 allele based on lipid and inflammatory marker profiles. Conclusions This study provides strong evidence linking APOE gene polymorphism to abnormal serum lipid and inflammatory profiles. Individuals carrying the ε4 alleles exhibited dysregulated lipid metabolism and abnormal inflammatory markers, increasing their risk of CVD and AD. Early detection and prompt diagnosis are crucial for implementing therapeutic, dietary, and lifestyle interventions to mitigate risks and prevent or delay lipid and inflammation-related disorders.
Collapse
Affiliation(s)
| | | | | | | | | | - Karthik Krishna
- Research & Development, Vibrant Sciences LLC, San Carlos, USA
| | - Qi Song
- Data Acquisition and Analysis, Vibrant America LLC, San Carlos, USA
| | - Tianhao Wang
- Data Acquisition and Analysis, Vibrant Sciences LLC, San Carlos, USA
| | - Kang Bei
- Data Acquisition and Analysis, Vibrant Sciences LLC, San Carlos, USA
| | | |
Collapse
|
3
|
Marco-Benedí V, Cenarro A, Laclaustra M, Calmarza P, Bea AM, Vila À, Morillas-Ariño C, Puzo J, Mediavilla Garcia JD, Fernández Alamán AI, Suárez Tembra M, Civeira F. Influence of triglyceride concentration in lipoprotein (a) as a function of dyslipidemia. CLINICA E INVESTIGACION EN ARTERIOSCLEROSIS : PUBLICACION OFICIAL DE LA SOCIEDAD ESPANOLA DE ARTERIOSCLEROSIS 2024; 36:71-77. [PMID: 38161102 DOI: 10.1016/j.arteri.2023.11.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/07/2023] [Revised: 11/29/2023] [Accepted: 11/30/2023] [Indexed: 01/03/2024]
Abstract
BACKGROUND Recently, an inverse relationship between the blood concentration of lipoprotein(a) (Lp(a)) and triglycerides (TG) has been demonstrated. The larger the VLDL particle size, the greater the presence of VLDL rich in apoliprotein E and in subjects with the apoE2/E2 genotype, the lower Lp(a) concentration. The mechanism of this inverse association is unknown. The objective of this analysis was to evaluate the Lp(a)-TG association in patients treated at the lipid units included in the registry of the Spanish Society of Atherosclerosis (SEA) by comparing the different dyslipidemias. PATIENTS AND METHODS Five thousand two hundred and seventy-five subjects ≥18 years of age registered in the registry before March 31, 2023, with Lp(a) concentration data and complete lipid profile information without treatment were included. RESULTS The mean age was 53.0 ± 14.0 years, with 48% women. The 9.5% of subjects (n = 502) had diabetes and the 22.4% (n = 1184) were obese. The median TG level was 130 mg/dL (IQR 88.0-210) and Lp(a) 55.0 nmol/L (IQR 17.9-156). Lp(a) concentration showed a negative association with TG concentration when TG values exceeded 300 mg/dL. Subjects with TG > 1000 mg/dL showed the lowest level of Lp(a), 17.9 nmol/L, and subjects with TG < 300 mg/dL had a mean Lp(a) concentration of 60.1 nmol/L. In subjects without diabetes or obesity, the inverse association of Lp(a)-TG was especially important (p < 0.001). The median Lp(a) was 58.3 nmol/L in those with TG < 300 mg/dL and 22.0 nmol/L if TG > 1000 mg/dL. No association was found between TG and Lp(a) in subjects with diabetes and obesity, nor in subjects with familial hypercholesterolemia. In subjects with multifactorial combined hyperlipemia with TG < 300 mg/dL, Lp(a) was 64.6 nmol/L; in the range of 300-399 mg/dL of TG, Lp(a) decreased to 38. 8 nmol/L, and up to 22.3 nmol/L when TG > 1000 mg/dL. CONCLUSIONS Our results show an inverse Lp(a)-TG relationship in TG concentrations > 300 mg/dL in subjects without diabetes, obesity and without familial hypercholesterolemia. Our results suggest that, in those hypertriglyceridemias due to hepatic overproduction of VLDL, the formation of Lp(a) is reduced, unlike those in which the peripheral catabolism of TG-rich lipoproteins is reduced.
Collapse
Affiliation(s)
- Victoria Marco-Benedí
- Hospital Universitario Miguel Servet, Instituto de Investigación Sanitaria Aragón (IIS Aragón), CIBERCV, Zaragoza, España; Universidad de Zaragoza, Zaragoza, España.
| | - Ana Cenarro
- Hospital Universitario Miguel Servet, Instituto de Investigación Sanitaria Aragón (IIS Aragón), CIBERCV, Zaragoza, España; Instituto Aragonés de Ciencias de la Salud (IACS), Zaragoza, España
| | - Martín Laclaustra
- Hospital Universitario Miguel Servet, Instituto de Investigación Sanitaria Aragón (IIS Aragón), CIBERCV, Zaragoza, España; Universidad de Zaragoza, Zaragoza, España
| | - Pilar Calmarza
- Hospital Universitario Miguel Servet, Instituto de Investigación Sanitaria Aragón (IIS Aragón), CIBERCV, Zaragoza, España
| | - Ana M Bea
- Hospital Universitario Miguel Servet, Instituto de Investigación Sanitaria Aragón (IIS Aragón), CIBERCV, Zaragoza, España
| | - Àlex Vila
- Unidad de Lípidos, Servicio de Medicina Interna, Hospital de Figueres, Figueres, España
| | - Carlos Morillas-Ariño
- Sección de Endocrinología y Nutrición, Hospital Universitario Dr. Peset, Valencia, España; Departamento de Medicina, Universidad de Valencia, Valencia, España
| | - José Puzo
- Unidad de Lípidos, Servicio de Análisis y Bioquímica Clínica, Hospital San Jorge, Huesca, España
| | | | | | - Manuel Suárez Tembra
- Unidad de Lípidos y Riesgo Cardiovascular, Servicio de Medicina Interna, Hospital San Rafael, A Coruña, España
| | - Fernando Civeira
- Hospital Universitario Miguel Servet, Instituto de Investigación Sanitaria Aragón (IIS Aragón), CIBERCV, Zaragoza, España; Universidad de Zaragoza, Zaragoza, España
| |
Collapse
|
4
|
Reijnders E, van der Laarse A, Ruhaak LR, Cobbaert CM. Closing the gaps in patient management of dyslipidemia: stepping into cardiovascular precision diagnostics with apolipoprotein profiling. Clin Proteomics 2024; 21:19. [PMID: 38429638 PMCID: PMC10908091 DOI: 10.1186/s12014-024-09465-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Accepted: 02/14/2024] [Indexed: 03/03/2024] Open
Abstract
In persons with dyslipidemia, a high residual risk of cardiovascular disease remains despite lipid lowering therapy. Current cardiovascular risk prediction mainly focuses on low-density lipoprotein cholesterol (LDL-c) levels, neglecting other contributing risk factors. Moreover, the efficacy of LDL-c lowering by statins resulting in reduced cardiovascular risk is only partially effective. Secondly, from a metrological viewpoint LDL-c falls short as a reliable measurand. Both direct and calculated LDL-c tests produce inaccurate test results at the low end under aggressive lipid lowering therapy. As LDL-c tests underperform both clinically and metrologically, there is an urging need for molecularly defined biomarkers. Over the years, apolipoproteins have emerged as promising biomarkers in the context of cardiovascular disease as they are the functional workhorses in lipid metabolism. Among these, apolipoprotein B (ApoB), present on all atherogenic lipoprotein particles, has demonstrated to clinically outperform LDL-c. Other apolipoproteins, such as Apo(a) - the characteristic apolipoprotein of the emerging risk factor lipoprotein(a) -, and ApoC-III - an inhibitor of triglyceride-rich lipoprotein clearance -, have attracted attention as well. To support personalized medicine, we need to move to molecularly defined risk markers, like the apolipoproteins. Molecularly defined diagnosis and molecularly targeted therapy require molecularly measured biomarkers. This review provides a summary of the scientific validity and (patho)physiological role of nine serum apolipoproteins, Apo(a), ApoB, ApoC-I, ApoC-II, ApoC-III, ApoE and its phenotypes, ApoA-I, ApoA-II, and ApoA-IV, in lipid metabolism, their association with cardiovascular disease, and their potential as cardiovascular risk markers when measured in a multiplex apolipoprotein panel.
Collapse
Affiliation(s)
- Esther Reijnders
- Department of Clinical Chemistry and Laboratory Medicine, Leiden University Medical Center, Leiden, the Netherlands.
| | - Arnoud van der Laarse
- Department of Clinical Chemistry and Laboratory Medicine, Leiden University Medical Center, Leiden, the Netherlands
| | - L Renee Ruhaak
- Department of Clinical Chemistry and Laboratory Medicine, Leiden University Medical Center, Leiden, the Netherlands
| | - Christa M Cobbaert
- Department of Clinical Chemistry and Laboratory Medicine, Leiden University Medical Center, Leiden, the Netherlands
| |
Collapse
|
5
|
Chen N, Jiang F, Chen X, Zhu L, Qiao N, Zhou J, Zhang Y. Associations of Lipoprotein(a) Level with Cerebral Small Vessel Disease in Patients with Alzheimer's Disease. Brain Sci 2023; 14:34. [PMID: 38248249 PMCID: PMC10813431 DOI: 10.3390/brainsci14010034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Revised: 12/25/2023] [Accepted: 12/27/2023] [Indexed: 01/23/2024] Open
Abstract
BACKGROUND This study aimed to examine the association of lipoprotein(a) [Lp(a)] level with the burden of cerebral small vessel disease (CSVD) in patients with Alzheimer's disease (AD). METHODS Data from 111 consecutive patients with AD admitted to Nanjing First Hospital from 2015 to 2022 were retrospectively analyzed in this study. Serum Lp(a) concentrations were grouped into tertiles (T1-T3). Brain magnetic resonance imaging (MRI) was rated for the presence of CSVD, including enlarged perivascular spaces (EPVS), lacunes, white-matter lesions, and cerebral microbleeds (CMBs). The CSVD burden was calculated by summing the scores of each MRI marker at baseline. A binary or ordinal logistic regression model was used to estimate the relationship of serum Lp(a) levels with CSVD burden and each MRI marker. RESULTS Patients with higher tertiles of Lp(a) levels were less likely to have any CSVD (T1, 94.6%; T2, 78.4%; T3, 66.2%; p = 0.013). Multivariable analysis found that Lp(a) levels were inversely associated with the presence of CSVD (T2 vs. T1: adjusted odds ratio [aOR] 0.132, 95% confidence interval [CI] 0.018-0.946, p = 0.044; T3 vs. T1: aOR 0.109, 95% CI 0.016-0.737, p = 0.023) and CSVD burden (T3 vs. T1: aOR 0.576, 95% CI 0.362-0.915, p = 0.019). The independent relationship between Lp(a) levels and individual CSVD features was significant for moderate-to-severe EPVS in the centrum semiovale (T2 vs. T1: aOR 0.059, 95% CI 0.006-0.542, p = 0.012; T3 vs. T1: aOR 0.029, 95% CI 0.003-0.273, p = 0.002) and CMBs (T3 vs. T1: aOR 0.144, 95% CI 0.029-0.716, p = 0.018). CONCLUSIONS In this study, serum Lp(a) level was inversely associated with CSVD in AD patients.
Collapse
Affiliation(s)
- Nihong Chen
- Department of Neurology, Nanjing First Hospital, Nanjing Medical University, Nanjing 210006, China; (N.C.); (X.C.); (L.Z.); (J.Z.)
- Department of Neurology, Nanjing Yuhua Hospital, Yuhua Branch of Nanjing First Hospital, Nanjing 210039, China;
| | - Fuping Jiang
- Department of Geriatrics, Nanjing First Hospital, Nanjing Medical University, Nanjing 210006, China;
| | - Xiangliang Chen
- Department of Neurology, Nanjing First Hospital, Nanjing Medical University, Nanjing 210006, China; (N.C.); (X.C.); (L.Z.); (J.Z.)
| | - Lin Zhu
- Department of Neurology, Nanjing First Hospital, Nanjing Medical University, Nanjing 210006, China; (N.C.); (X.C.); (L.Z.); (J.Z.)
| | - Na Qiao
- Department of Neurology, Nanjing Yuhua Hospital, Yuhua Branch of Nanjing First Hospital, Nanjing 210039, China;
| | - Junshan Zhou
- Department of Neurology, Nanjing First Hospital, Nanjing Medical University, Nanjing 210006, China; (N.C.); (X.C.); (L.Z.); (J.Z.)
| | - Yingdong Zhang
- Department of Neurology, Nanjing First Hospital, Nanjing Medical University, Nanjing 210006, China; (N.C.); (X.C.); (L.Z.); (J.Z.)
| |
Collapse
|
6
|
Brandt EJ, Brandt DJ, Desai NR, Spatz ES, Nasir K, Mani A. Association of vitamins, minerals, and lead with lipoprotein(a) in a cross-sectional cohort of US adults. INT J VITAM NUTR RES 2023; 93:99-110. [PMID: 34024154 PMCID: PMC8964024 DOI: 10.1024/0300-9831/a000709] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Lipoprotein(a)(Lp[a]) is a low-density lipoprotein-cholesterol (LDL-C)-like particle with potent pro-atherothrombotic properties. The association of Lp(a) with several circulating factors, including vitamins, remains unresolved. We performed an observational analysis using the National Health and Nutrition Examination Survey III cohort, a cohort used to monitor the nutrition status of US-citizens. We used multivariable linear regression to test associations of Lp(a) and LDL-C with levels of serum vitamins and minerals and whole-blood lead. Analyses controlled for factors known to associate with Lp(a) (age, sex, race/ethnicity, statin use, hemoglobin A1c, body mass index, hypertension, diabetes, glomerular filtration rate, alcohol intake, and saturated fat intake). LDL-C was corrected for Lp(a) mass. Multiple sensitivity tests were performed, including considering factors as categorical variables (deficient, normal, elevated). Among 7,662 subjects, Lp(a) correlated (β-coefficient) positively (change per 1 conventional unit increase) with carotenoids (lycopene (0.17(0.06,0.28), p=0.005), lutein (0.19(0.07,0.30), p=0.002), β-cryptoxanthin (0.21(0.05,0.37), p=0.01), β-carotene (0.05(0.02,0.09), p=0.003), and α-carotene (0.15(0.01,0.30), p=0.04)) and lead (0.54(0.03,1.05), p=0.04) levels when tested as continuous variables. LDL-C had similar associations. Lp(a) did not associate with vitamins A, B12, C, or E retinyl esters, folate, RBC-folate, selenium, ferritin, transferrin saturation, or calcium. With factors as categorical variables, Lp(a) but not LDL-C negatively associated with elevated vitamin B12 (-5.41(-9.50, -1.53), p=0.01) and folate (-2.86(-5.09, -0.63), p=0.01). In conclusion, Lp(a) associated similarly to LDL-C when vitamins, minerals, and lead were tested as continuous variables, while only Lp(a) correlated with vitamin B12 and folate when tested as categorical variables. These observations are hypotheses generating and require further studies to determine causality.
Collapse
Affiliation(s)
- Eric J. Brandt
- National Clinician Scholars Program, Yale School of Medicine, New Haven, CT, USA
- Division of Cardiovascular Medicine, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Daniel J. Brandt
- Department of Epidemiology, University of Michigan School of Public Health, Ann Arbor, MI, USA
| | - Nihar R. Desai
- Section of Cardiovascular Medicine, Department of Internal Medicine, Yale School of Medicine, New Haven, CT, USA
- Center for Outcomes Research and Evaluation, Yale-New Haven Hospital, New Haven, CT, USA
| | - Erica S. Spatz
- Section of Cardiovascular Medicine, Department of Internal Medicine, Yale School of Medicine, New Haven, CT, USA
- Center for Outcomes Research and Evaluation, Yale-New Haven Hospital, New Haven, CT, USA
| | - Khurram Nasir
- Center for Outcomes Research, Houston Methodist DeBakey Heart and Vascular Center, Houston, TX, USA
| | - Arya Mani
- Section of Cardiovascular Medicine, Department of Internal Medicine, Yale School of Medicine, New Haven, CT, USA
- Department of Genetics, Yale School of Medicine, New Haven, CT, USA
| |
Collapse
|
7
|
Ying Q, Croyal M, Chan DC, Blanchard V, Pang J, Krempf M, Watts GF. Effect of Omega-3 Fatty Acid Supplementation on the Postprandial Metabolism of Apolipoprotein(a) in Familial Hypercholesterolemia. J Atheroscler Thromb 2023; 30:274-286. [PMID: 35676030 PMCID: PMC9981347 DOI: 10.5551/jat.63587] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Abstract
AIM Lipoprotein(a) (Lp(a)) is a low-density lipoprotein-like particle containing apolipoprotein(a) (apo(a)) that increases the risk of atherosclerotic cardiovascular disease (ASCVD) in familial hypercholesterolemia (FH). Postprandial redistribution of apo(a) protein from Lp(a) to triglyceride-rich lipoproteins (TRLs) may also increase the atherogenicity of TRL particles. Omega-3 fatty acid (ω3FA) supplementation improves postprandial TRL metabolism in FH subjects. However, its effect on postprandial apo(a) metabolism has yet to be investigated. METHODS We carried out an 8-week open-label, randomized, crossover trial to test the effect of ω3FA supplementation (4 g/day) on postprandial apo(a) responses in FH patients following ingestion of an oral fat load. Postprandial plasma total and TRL-apo(a) concentrations were measured by liquid chromatography with tandem mass spectrometry, and the corresponding areas under the curve (AUCs) (0-10h) were determined using the trapezium rule. RESULTS Compared with no ω3FA treatment, ω3FA supplementation significantly lowered the concentrations of postprandial TRL-apo(a) at 0.5 (-17.9%), 1 (-18.7%), 2 (-32.6%), and 3 h (-19.2%) (P<0.05 for all). Postprandial TRL-apo(a) AUC was significantly reduced with ω3FA by 14.8% (P<0.05). By contrast, ω3FA had no significant effect on the total AUCs of apo(a), apoC-III, and apoE (P>0.05 for all). The decrease in postprandial TRL-apo(a) AUC was significantly associated with changes in the AUC of triglycerides (r=0.600; P<0.01) and apoB-48 (r=0.616; P<0.01). CONCLUSIONS Supplementation with ω3FA reduces postprandial TRL-apo(a) response to a fat meal in FH patients; this novel metabolic effect of ω3FA may have implications on decreasing the risk of ASCVD in patients with FH, especially in those with elevated plasma triglyceride and Lp(a) concentrations. However, the clinical implications of these metabolic findings require further evaluation in outcome or surrogate endpoint trials.
Collapse
Affiliation(s)
- Qidi Ying
- Medical School, University of Western Australia, Perth, Western Australia, Australia
| | - Mikaël Croyal
- Nantes Universite, CNRS, INSERM, l’institut du thorax, F-44000 Nantes, France,Nantes Universite, CHU Nantes, INSERM, CNRS, SFR Sante, INSERM UMS 016, CNRS UMS 3556, F-44000 Nantes, France,CRNH-Ouest Mass Spectrometry Core Facility, F-44000 Nantes, France
| | - Dick C Chan
- Medical School, University of Western Australia, Perth, Western Australia, Australia
| | - Valentin Blanchard
- Department of Medicine, Centre for Heart Lung Innovation, Providence Healthcare Research Institute, St. Paul’s Hospital, University of British Columbia, Vancouver, Canada
| | - Jing Pang
- Medical School, University of Western Australia, Perth, Western Australia, Australia
| | | | - Gerald F Watts
- Medical School, University of Western Australia, Perth, Western Australia, Australia,Lipid Disorders Clinic, Department of Cardiology and Internal Medicine, Royal Perth Hospital, Perth, Western Australia, Australia
| |
Collapse
|
8
|
Bea AM, Cenarro A, Marco-Bened V, Laclaustra M, Martn C, Ibarretxe D, Pint X, Arrobas T, Vials C, Civeira F, Olmos S. Diagnosis of Familial Dysbetalipoproteinemia Based on the Lipid Abnormalities Driven by APOE2/E2 Genotype. Clin Chem 2023; 69:140-148. [PMID: 36644927 DOI: 10.1093/clinchem/hvac213] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Accepted: 11/14/2022] [Indexed: 01/17/2023]
Abstract
BACKGROUND Familial dysbetalipoproteinemia (FDBL) is a monogenic disease due to variants in APOE with a highly variable phenotype. Current diagnostic lipid-based methods have important limitations. The objective is twofold: to define characteristics of dysbetalipoproteinemia (DBL) based on the analysis of APOE in patients from a lipid unit and in a sample from the general population, and to propose a screening algorithm for FDBL. METHODS Lipids and APOE genotype from consecutive unrelated subjects from Miguel Servet University Hospital (MSUH) (n 3603), subjects from the general population participants of the Aragon Workers Health Study (AWHS) (n 4981), and selected subjects from external lipid units (Ext) (n 390) were used to define DBL criteria and to train and validate a screening tool. RESULTS Thirty-five subjects from MSUH, 21 subjects from AWHS, and 31 subjects from Ext were APOE2/2 homozygous. The combination of non high-density lipoprotein cholesterol (non-HDLc)/apoB 1.7 plus triglycerides/apoB 1.35, in mg/dL (non-HDLc [mmol/L]/apolipoprotein B (apoB) [g/L] 4.4 and triglycerides [mmol/L]/apoB [g/L] 3.5), provided the best diagnostic performance for the identification of subjects with hyperlipidemia and APOE2/2 genotype (sensitivity 100 in the 3 cohorts, and specificity 92.8 [MSUH], 80.9 [AWHS], and 77.6 [Ext]). This improves the performance of previous algorithms. Similar sensitivity and specificity were observed in APOE2/2 subjects receiving lipid-lowering drugs. CONCLUSIONS The combination of non-HDLc/apoB and triglycerides/apoB ratios is a valuable tool to diagnose DBL in patients with hyperlipidemia with or without lipid-lowering drugs. FDBL diagnosis requires DBL and the presence of a compatible APOE genotype. Most adult APOE2/2 subjects express DBL, making FDBL as common as familial hypercholesterolemia in the population.
Collapse
Affiliation(s)
- Ana M Bea
- Hospital Universitario Miguel Servet, IIS Aragn, CIBERCV, Zaragoza, Spain
| | - Ana Cenarro
- Hospital Universitario Miguel Servet, IIS Aragn, CIBERCV, Zaragoza, Spain.,Molecular Research Laboratory, Instituto Aragones de Ciencias de la Salud (IACS), Zaragoza, Spain
| | - Victoria Marco-Bened
- Hospital Universitario Miguel Servet, IIS Aragn, CIBERCV, Zaragoza, Spain.,Departamento de Medicina, Psiquiatra y Dermatologa, Universidad de Zaragoza, Zaragoza, Spain
| | - Martn Laclaustra
- Hospital Universitario Miguel Servet, IIS Aragn, CIBERCV, Zaragoza, Spain.,Departamento de Medicina, Psiquiatra y Dermatologa, Universidad de Zaragoza, Zaragoza, Spain
| | - Csar Martn
- Fundacin Biofisika Bizkaia, Leioa, Spain.,Biofisika Institute (UPV/EHU, CSIC), Leioa, Spain.,Department of Biochemistry and Molecular Biology, Universidad del Pas Vasco UPV/EHU, Bilbao, Spain
| | - Daiana Ibarretxe
- Unitat de Medicina Vascular i Metabolisme (UVASMET) Hospital Universitari Sant Joan, IISPV, CIBERDEM, Universitat Rovira i Virgili, Reus, Tarragona, Spain
| | - Xavier Pint
- Unidad de Lpidos, Servicio de Medicina Interna, Hospital Universitario de Bellvitge-Idibell, Universidad de Barcelona, CiberObn, Barcelona, Spain
| | - Teresa Arrobas
- Laboratorio de Nutricin y RCV, Laboratorio de Bioqumica Clnica, Hospital Virgen Macarena, Sevilla, Spain
| | - Clara Vials
- Endocrinology Department, Hospital Clnic de Barcelona, Barcelona, Spain.,Institut dInvestigacions Biomdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain.,Centro de Investigacin Biomdica en Red Fisiopatologa de la Obesidad y Nutricin (CIBEROBN), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| | - Fernando Civeira
- Hospital Universitario Miguel Servet, IIS Aragn, CIBERCV, Zaragoza, Spain.,Departamento de Medicina, Psiquiatra y Dermatologa, Universidad de Zaragoza, Zaragoza, Spain
| | - Salvador Olmos
- Hospital Universitario Miguel Servet, IIS Aragn, CIBERCV, Zaragoza, Spain.,Aragon Institute of Engineering Research (I3A), Universidad de Zaragoza, Zaragoza, Spain
| |
Collapse
|
9
|
Nestel P, Loh WJ, Ward NC, Watts GF. New Horizons: Revival of Lipoprotein (a) as a Risk Factor for Cardiovascular Disease. J Clin Endocrinol Metab 2022; 107:e4281-e4294. [PMID: 36108076 DOI: 10.1210/clinem/dgac541] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/09/2022] [Indexed: 02/13/2023]
Abstract
The status of lipoprotein (a) [Lp(a)] as a cardiovascular risk factor has been resurrected by advances in genetics. Mendelian randomization studies show a causal link of Lp(a) with coronary artery disease (CAD), peripheral artery disease (PAD), and calcific aortic valve stenosis (CAVS). The genetics of Lp(a) is complex and extends beyond the kringle-IV type 2, as it is also dependent on ancestry. The plasma concentration of Lp(a) is determined by the hepatic production of apolipoprotein(a) [apo(a)] component of Lp(a), supporting the use of nucleic acids that inhibit the messenger RNA (mRNA) gene transcript for apo(a). Analytical barriers to measurement of Lp(a) are being addressed using isoform independent assays and a traceable standard. The association of Lp(a) and atherosclerotic cardiovascular disease is higher for myocardial infarction than PAD and CAVS. Increased risk of type 2 diabetes mellitus associated with low Lp(a) levels is perplexing and requires further investigation. The greatest advancement in Lp(a)-lowering therapies is based on using RNA therapeutics that are now being investigated in clinical trials. Proprotein convertase subtilisin/kexin type 9 (PCSK9) inhibition lowers Lp(a) modestly, but whether cardiovascular benefit is independent of low-density lipoprotein lowering remains unclear. Opportunistic and selective testing for Lp(a) is supported by moderate evidence, with the case for universal screening premature. Modification of behavioral and clinical risk factors may be targeted to mitigate Lp(a)-mediated risk of cardiovascular disease. Clinical practice guidelines have been developed to address gaps in care of high Lp(a), but full implementation awaits the findings of clinical outcome trials using RNA-directed therapies currently underway.
Collapse
Affiliation(s)
- Paul Nestel
- Baker Heart and Diabetes Institute, Melbourne, Australia
| | - Wann Jia Loh
- School of Medicine, University of Western Australia, Perth, Australia
- Department of Cardiology and Internal Medicine, Royal Perth Hospital, Perth, Australia
- Department of Endocrinology, Changi General Hospital, Singapore
- Duke-NUS Medical School, Singapore
| | - Natalie C Ward
- School of Medicine, University of Western Australia, Perth, Australia
| | - Gerald F Watts
- School of Medicine, University of Western Australia, Perth, Australia
- Department of Cardiology and Internal Medicine, Royal Perth Hospital, Perth, Australia
| |
Collapse
|
10
|
Cui T, El Mekkaoui K, Reinvall J, Havulinna AS, Marttinen P, Kaski S. Gene-gene interaction detection with deep learning. Commun Biol 2022; 5:1238. [PMID: 36371468 PMCID: PMC9653457 DOI: 10.1038/s42003-022-04186-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2022] [Accepted: 10/27/2022] [Indexed: 11/13/2022] Open
Abstract
The extent to which genetic interactions affect observed phenotypes is generally unknown because current interaction detection approaches only consider simple interactions between top SNPs of genes. We introduce an open-source framework for increasing the power of interaction detection by considering all SNPs within a selected set of genes and complex interactions between them, beyond only the currently considered multiplicative relationships. In brief, the relation between SNPs and a phenotype is captured by a neural network, and the interactions are quantified by Shapley scores between hidden nodes, which are gene representations that optimally combine information from the corresponding SNPs. Additionally, we design a permutation procedure tailored for neural networks to assess the significance of interactions, which outperformed existing alternatives on simulated datasets with complex interactions, and in a cholesterol study on the UK Biobank it detected nine interactions which replicated on an independent FINRISK dataset.
Collapse
Affiliation(s)
- Tianyu Cui
- Department of Computer Science, Aalto University, Espoo, Finland.
| | | | - Jaakko Reinvall
- Department of Computer Science, Aalto University, Espoo, Finland
| | - Aki S Havulinna
- Finnish Institute for Health and Welfare (THL), Helsinki, Finland
- Institute for Molecular Medicine Finland, FIMM-HiLIFE, Helsinki, Finland
| | - Pekka Marttinen
- Department of Computer Science, Aalto University, Espoo, Finland
- Finnish Institute for Health and Welfare (THL), Helsinki, Finland
| | - Samuel Kaski
- Department of Computer Science, Aalto University, Espoo, Finland
- Department of Computer Science, University of Manchester, Manchester, UK
| |
Collapse
|
11
|
Ramos-Cáceres M, Lamiquiz-Moneo I, Cenarro A, Calmarza P, Marco-Benedí V, Bea AM, Mateo-Gallego R, Puzo J, Ordovas JM, Civeira F, Laclaustra M. Triglyceride Metabolism Modifies Lipoprotein(a) Plasma Concentration. J Clin Endocrinol Metab 2022; 107:e3594-e3602. [PMID: 35789387 DOI: 10.1210/clinem/dgac412] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/24/2022] [Indexed: 11/19/2022]
Abstract
BACKGROUND Lipoprotein(a) (Lp(a)) is a significant cardiovascular risk factor. Knowing the mechanisms that regulate its concentration can facilitate the development of Lp(a)-lowering drugs. This study analyzes the relationship between triglycerides (TGs) and Lp(a) concentrations, cross-sectionally and longitudinally, and the influence of the number and composition of TG-rich lipoproteins, and the APOE genotype. METHODS Data from Aragon Workers Health Study (AWHS) (n = 5467), National Health and Nutrition Examination Survey III phase 2 (n = 3860), and Hospital Universitario Miguel Servet (HUMS) (n = 2079) were used for cross-sectional TG and Lp(a) relationship. Lp(a) intrasubject variation was studied in AWHS participants and HUMS patients with repeated measurements. TG-rich lipoproteins were quantified by nuclear magnetic resonance in a subsample from AWHS. Apolipoproteins B and E were quantified by Luminex in very low-density lipoprotein (VLDL) isolated by ultracentrifugation, from HUMS samples. APOE genotyping was carried in AWHS and HUMS participants. Regression models adjusted for age and sex were used to study the association. RESULTS The 3 studies showed an inverse relationship between TG and Lp(a). Increased VLDL number, size, and TG content were associated with significantly lower Lp(a). There was an inverse association between the apoE concentration in VLDL and Lp(a). No significant association was observed for apolipoprotein (apo)B. Subjects carrying the apoE2/E2 genotype had significantly lower levels of Lp(a). CONCLUSION Our results show an inverse relationship Lp(a)-TG. Subjects with larger VLDL size have lower Lp(a), and lower values of Lp(a) were present in patients with apoE-rich VLDL and apoE2/E2 subjects. Our results suggest that bigger VLDLs and VLDLs enriched in apoE are inversely involved in Lp(a) plasma concentration.
Collapse
Affiliation(s)
- Maria Ramos-Cáceres
- Hospital Universitario Miguel Servet, Instituto de Investigación Sanitaria Aragón (IIS Aragón), CIBERCV, Zaragoza 50009, Spain
| | - Itziar Lamiquiz-Moneo
- Hospital Universitario Miguel Servet, Instituto de Investigación Sanitaria Aragón (IIS Aragón), CIBERCV, Zaragoza 50009, Spain
- Departamento de Anatomía e Histología Humanas, Facultad de Medicina, Universidad de Zaragoza, Zaragoza 50009, Spain
| | - Ana Cenarro
- Hospital Universitario Miguel Servet, Instituto de Investigación Sanitaria Aragón (IIS Aragón), CIBERCV, Zaragoza 50009, Spain
- Instituto Aragonés de Ciencias de la Salud, (IACS), Zaragoza 50009, Spain
| | - Pilar Calmarza
- Hospital Universitario Miguel Servet, Instituto de Investigación Sanitaria Aragón (IIS Aragón), CIBERCV, Zaragoza 50009, Spain
| | - Victoria Marco-Benedí
- Hospital Universitario Miguel Servet, Instituto de Investigación Sanitaria Aragón (IIS Aragón), CIBERCV, Zaragoza 50009, Spain
- Departamento de Medicina, Psiquiatría y Dermatología, Facultad de Medicina, Universidad de Zaragoza, Zaragoza 50009, Spain
| | - Ana M Bea
- Hospital Universitario Miguel Servet, Instituto de Investigación Sanitaria Aragón (IIS Aragón), CIBERCV, Zaragoza 50009, Spain
| | - Rocio Mateo-Gallego
- Hospital Universitario Miguel Servet, Instituto de Investigación Sanitaria Aragón (IIS Aragón), CIBERCV, Zaragoza 50009, Spain
- Departamento de Fisiatría y Enfermería, Facultad de Ciencias de la Salud y del Deporte, Universidad de Zaragoza, Huesca 22002, Spain
| | - Jose Puzo
- Departamento de Medicina, Psiquiatría y Dermatología, Facultad de Medicina, Universidad de Zaragoza, Zaragoza 50009, Spain
- Unidad de Lípidos, Servicio de Análisis y Bioquímica Clínica, Hospital San Jorge, Huesca 22004, Spain
| | - Jose M Ordovas
- Nutrition and Genomics Laboratory, JM-USDA Human Nutrition Research Center on Aging at Tufts University, Boston, Massachusetts 02111, USA
- Precision Nutrition and Obesity Program, IMDEA Alimentación, Madrid 28049, Spain
| | - Fernando Civeira
- Hospital Universitario Miguel Servet, Instituto de Investigación Sanitaria Aragón (IIS Aragón), CIBERCV, Zaragoza 50009, Spain
- Departamento de Medicina, Psiquiatría y Dermatología, Facultad de Medicina, Universidad de Zaragoza, Zaragoza 50009, Spain
| | - Martin Laclaustra
- Hospital Universitario Miguel Servet, Instituto de Investigación Sanitaria Aragón (IIS Aragón), CIBERCV, Zaragoza 50009, Spain
- Departamento de Medicina, Psiquiatría y Dermatología, Facultad de Medicina, Universidad de Zaragoza, Zaragoza 50009, Spain
| |
Collapse
|
12
|
Durrington PN, Bashir B, Bhatnagar D, Soran H. Lipoprotein (a) in familial hypercholesterolaemia. Curr Opin Lipidol 2022; 33:257-263. [PMID: 35942820 DOI: 10.1097/mol.0000000000000839] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
PURPOSE OF REVIEW The role of lipoprotein (a) in atherogenesis has been the subject of argument for many years. Evidence that it is raised in familial hypercholesterolaemia has been disputed not least because a mechanism related to low density lipoprotein (LDL) receptor mediated catabolism has been lacking. Whether lipoprotein (a) increases the already raised atherosclerotic cardiovascular disease (ASCVD) risk in familial hypercholesterolaemia is also more dubious than is often stated. We review the evidence in an attempt to provide greater clarity. RECENT FINDINGS Lipoprotein (a) levels are raised as a consequence of inheriting familial hypercholesterolaemia. The mechanism for this is likely to involve increased hepatic production, probably mediated by PCSK9 augmented by apolipoprotein E. The extent to which raised lipoprotein (a) contributes to the increased ASCVD risk in familial hypercholesterolaemia remains controversial.Unlike, for example, statins which are effective across the whole spectrum of LDL concentrations, drugs in development to specifically lower lipoprotein (a) are likely to be most effective in people with the highest levels of lipoprotein (a). People with familial hypercholesterolaemia may therefore be in the vanguard of those in whom theses agents should be exhibited. SUMMARY Inheritance of familial hypercholesterolaemia undoubtedly increases the likelihood that lipoprotein (a) will be raised. However, in familial hypercholesterolaemia when ASCVD incidence is already greatly increased due to high LDL cholesterol, whether lipoprotein (a) contributes further to this risk cogently needs to be tested with drugs designed to specifically lower lipoprotein (a).
Collapse
Affiliation(s)
- Paul N Durrington
- Cardiovascular Research Group, Faculty of Biology, Medicine and Health, University of Manchester
| | - Bilal Bashir
- Cardiovascular Research Group, Faculty of Biology, Medicine and Health, University of Manchester
- Manchester National Institute for Health Research/Wellcome Trust Clinical Research Facility, Manchester
| | - Deepak Bhatnagar
- Cardiovascular Research Group, Faculty of Biology, Medicine and Health, University of Manchester
| | - Handrean Soran
- Cardiovascular Research Group, Faculty of Biology, Medicine and Health, University of Manchester
- Manchester National Institute for Health Research/Wellcome Trust Clinical Research Facility, Manchester
- Department of Diabetes, Endocrinology and Metabolism, Manchester University NHS Foundation Trust, Manchester, UK
| |
Collapse
|
13
|
Raitakari O, Kivelä A, Pahkala K, Rovio S, Mykkänen J, Ahola-Olli A, Loo BM, Lyytikäinen LP, Lehtimäki T, Kähönen M, Juonala M, Rönnemaa T, Lamina C, Kronenberg F, Viikari J. Long-term tracking and population characteristics of lipoprotein (a) in the cardiovascular risk in young finns study. Atherosclerosis 2022; 356:18-27. [DOI: 10.1016/j.atherosclerosis.2022.07.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Revised: 06/01/2022] [Accepted: 07/13/2022] [Indexed: 11/16/2022]
|
14
|
Loh WJ, Chan DC, Mata P, Watts GF. Familial Hypercholesterolemia and Elevated Lipoprotein(a): Cascade Testing and Other Implications for Contextual Models of Care. Front Genet 2022; 13:905941. [PMID: 35571022 PMCID: PMC9091303 DOI: 10.3389/fgene.2022.905941] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Accepted: 04/04/2022] [Indexed: 11/13/2022] Open
Abstract
Elevated lipoprotein(a) [Lp(a)], a predominantly genetic disorder, is a causal risk factor for atherosclerotic cardiovascular disease (ASCVD) and calcific aortic valvular disease, particularly in patients with familial hypercholesterolemia (FH), a Tier I genomic condition. The combination from birth of the cumulative exposure to elevated plasma concentrations of both Lp(a) and low-density lipoprotein is particularly detrimental and explains the enhanced morbidity and mortality risk observed in patients with both conditions. An excellent opportunity to identify at-risk patients with hyper-Lp(a) at increased risk of ASCVD is to test for hyper-Lp(a) during cascade testing for FH. With probands having FH and hyper-Lp(a), the yield of detection of hyper-Lp(a) is 1 individual for every 2.1-2.4 relatives tested, whereas the yield of detection of both conditions is 1 individual for every 3-3.4 relatives tested. In this article, we discuss the incorporation of assessment of Lp(a) in the cascade testing in FH as a feasible and crucial part of models of care for FH. We also propose a simple management tool to help physicians identify and manage elevated Lp(a) in FH, with implications for the care of Lp(a) beyond FH, noting that the clinical use of RNA therapeutics for specifically targeting the overproduction of Lp(a) in at risk patients is still under investigation.
Collapse
Affiliation(s)
- Wann Jia Loh
- Department of Endocrinology, Changi General Hospital, Singapore, Singapore
| | - Dick C Chan
- Medical School, University of Western Australia, Perth, WA, Australia
| | - Pedro Mata
- Fundación Hipercolesterolemia Familiar, Madrid, Spain
| | - Gerald F Watts
- Medical School, University of Western Australia, Perth, WA, Australia.,Lipid Disorders Clinic, Department of Cardiology and Internal Medicine, Royal Perth Hospital, Perth, WA, Australia
| |
Collapse
|
15
|
Chemello K, Blom DJ, Marais AD, Lambert G, Blanchard V. Genetic and Mechanistic Insights into the Modulation of Circulating Lipoprotein (a) Concentration by Apolipoprotein E Isoforms. Curr Atheroscler Rep 2022; 24:399-405. [PMID: 35355214 DOI: 10.1007/s11883-022-01016-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/04/2022] [Indexed: 01/04/2023]
Abstract
PURPOSE OF REVIEW Lipoprotein (a) [Lp(a)] is a highly atherogenic lipoprotein species. A unique feature of Lp(a) is the strong genetic determination of its concentration. The LPA gene is responsible for up to 90% of the variance in Lp(a), but other genes also have an impact. RECENT FINDINGS Genome-wide associations studies indicate that the APOE gene, encoding apolipoprotein E (apoE), is the second most important locus modulating Lp(a) concentrations. Population studies clearly show that carriers of the apoE2 variant (ε2) display reduced Lp(a) levels, the lowest concentrations being observed in ε2/ε2 homozygotes. This genotype can lead predisposed adults to develop dysbetalipoproteinemia, a lipid disorder characterized by sharp elevations in cholesterol and triglycerides. However, dysbetalipoproteinemia does not significantly modulate circulating Lp(a). Mechanistically, apoE appears to impair the production but not the catabolism of Lp(a). These observations underline the complexity of Lp(a) metabolism and provide key insights into the pathways governing Lp(a) synthesis and secretion.
Collapse
Affiliation(s)
- Kévin Chemello
- Laboratoire Inserm, UMR 1188 DéTROI, Université de La Réunion, 2 Rue Maxime Rivière, 97490, Sainte Clotilde, France
| | - Dirk J Blom
- Division of Lipidology and Cape Heart Institute, Department of Medicine, University of Cape Town, Cape Town, South Africa
| | - A David Marais
- Division of Chemical Pathology, Department of Pathology, University of Cape Town, Cape Town, South Africa
| | - Gilles Lambert
- Laboratoire Inserm, UMR 1188 DéTROI, Université de La Réunion, 2 Rue Maxime Rivière, 97490, Sainte Clotilde, France.
| | - Valentin Blanchard
- Laboratoire Inserm, UMR 1188 DéTROI, Université de La Réunion, 2 Rue Maxime Rivière, 97490, Sainte Clotilde, France.,Departments of Medicine, Centre for Heart Lung Innovation, Providence Healthcare Research Institute, St. Paul's Hospital, University of British Columbia, Vancouver, Canada
| |
Collapse
|
16
|
Yeang C, Karwatowska-Prokopczuk E, Su F, Dinh B, Xia S, Witztum JL, Tsimikas S. Effect of Pelacarsen on Lipoprotein(a) Cholesterol and Corrected Low-Density Lipoprotein Cholesterol. J Am Coll Cardiol 2022; 79:1035-1046. [PMID: 35300814 DOI: 10.1016/j.jacc.2021.12.032] [Citation(s) in RCA: 81] [Impact Index Per Article: 40.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Accepted: 12/17/2021] [Indexed: 12/20/2022]
Abstract
BACKGROUND Laboratory methods that report low-density lipoprotein cholesterol (LDL-C) include both LDL-C and lipoprotein(a) cholesterol [Lp(a)-C] content. OBJECTIVES The purpose of this study was to assess the effect of pelacarsen on directly measured Lp(a)-C and LDL-C corrected for its Lp(a)-C content. METHODS The authors evaluated subjects with a history of cardiovascular disease and elevated Lp(a) randomized to 5 groups of cumulative monthly doses of 20-80 mg pelacarsen vs placebo. Direct Lp(a)-C was measured on isolated Lp(a) using LPA4-magnetic beads directed to apolipoprotein(a). LDL-C was reported as: 1) LDL-C as reported by the clinical laboratory; 2) LDL-Ccorr = laboratory-reported LDL-C - direct Lp(a)-C; and 3) LDL-CcorrDahlén = laboratory LDL-C - [Lp(a) mass × 0.30] estimated by the Dahlén formula. RESULTS The baseline median Lp(a)-C values in the groups ranged from 11.9 to 15.6 mg/dL. Compared with placebo, pelacarsen resulted in dose-dependent decreases in Lp(a)-C (2% vs -29% to -67%; P = 0.001-<0.0001). Baseline laboratory-reported mean LDL-C ranged from 68.5 to 89.5 mg/dL, whereas LDL-Ccorr ranged from 55 to 74 mg/dL. Pelacarsen resulted in mean percent/absolute changes of -2% to -19%/-0.7 to -8.0 mg/dL (P = 0.95-0.05) in LDL-Ccorr, -7% to -26%/-5.4 to -9.4 mg/dL (P = 0.44-<0.0001) in laboratory-reported LDL-C, and 3.1% to 28.3%/0.1 to 9.5 mg/dL (P = 0.006-0.50) increases in LDL-CcorrDahlén. Total apoB declined by 3%-16% (P = 0.40-<0.0001), but non-Lp(a) apoB was not significantly changed. CONCLUSIONS Pelacarsen significantly lowers direct Lp(a)-C and has neutral to mild lowering of LDL-Ccorr. In patients with elevated Lp(a), LDL-Ccorr provides a more accurate reflection of changes in LDL-C than either laboratory-reported LDL-C or the Dahlén formula.
Collapse
Affiliation(s)
- Calvin Yeang
- Division of Cardiovascular Medicine, Department of Medicine, University of California-San Diego, La Jolla, California, USA
| | | | - Fei Su
- Division of Cardiovascular Medicine, Department of Medicine, University of California-San Diego, La Jolla, California, USA
| | - Brian Dinh
- Division of Cardiovascular Medicine, Department of Medicine, University of California-San Diego, La Jolla, California, USA
| | - Shuting Xia
- Ionis Pharmaceuticals, Carlsbad, California, USA
| | - Joseph L Witztum
- Division of Endocrinology and Metabolism, Department of Medicine, University of California-San Diego, La Jolla, California, USA
| | - Sotirios Tsimikas
- Division of Cardiovascular Medicine, Department of Medicine, University of California-San Diego, La Jolla, California, USA; Ionis Pharmaceuticals, Carlsbad, California, USA.
| |
Collapse
|
17
|
Loh WJ, Chang X, Aw TC, Phua SK, Low AF, Chan MYY, Watts GF, Heng CK. Lipoprotein(a) as predictor of coronary artery disease and myocardial infarction in a multi-ethnic Asian population. Atherosclerosis 2021; 349:160-165. [PMID: 34887076 DOI: 10.1016/j.atherosclerosis.2021.11.018] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Revised: 11/15/2021] [Accepted: 11/17/2021] [Indexed: 02/07/2023]
Abstract
BACKGROUND AND AIMS The role of Lp(a) in multi-ethnic Asian populations with coronary artery disease (CAD) has not been well established. The aims of this study were (i) to investigate whether Lp(a) is a predictor of CAD, and (ii) amongst patients with CAD, to ascertain whether Lp(a) is a predictor of acute myocardial infarction (AMI) and severity of CAD. METHODS We compared three cardiovascular phenotypes from patients recruited at coronary angiography. CAD was defined as ≥50% coronary artery stenosis and subdivided into a group with AMI history (CAD+AMI+) and a group without (CAD+AMI-). Minimal CAD group (CAD-) was defined as normal or <30% coronary artery stenosis and no AMI. The severity of CAD was defined using the modified Gensini score. RESULTS We studied 2025 patients comprising 94.5% men and 61.4% of Chinese ethnicity. The median Lp(a) level was highest in CAD+AMI+, followed by CAD+AMI- and CAD- (26.2, 20.1, and 15.8 nmol/L respectively). Similarly, the frequency of patients with Lp(a) ≥120 nmol/L were in the same order (11.8%, 9.1% and 2.4%). Lp(a) levels were highest among Asian Indians, followed by Malays and Chinese patients (p < 0.001). Lp(a) levels and Lp(a) ≥120 nmol/L were significant predictors of CAD (Odds ratio (OR) = 1.12 per 10 nmol/L increment, p < 0.001, and OR = 5.41 p = 0.004 respectively). Among patients with CAD, higher Lp(a) levels were associated with increased AMI risk (OR = 1.02 per 10 nmol/L increment, p = 0.024). Lp(a) ≥120 nmol/L was positively associated with CAD severity (p = 0.020). CONCLUSIONS Plasma Lp(a) concentration is a positive predictor of CAD and AMI in a mostly male South East Asian population.
Collapse
Affiliation(s)
- Wann Jia Loh
- Department of Endocrinology, Changi General Hospital, 2 Simei Street 3, 529889, Singapore.
| | - Xuling Chang
- Department of Paediatrics, Yong Loo Lin School of Medicine, National University of Singapore, 1E Kent Ridge Road, NUHS Tower Block, Level 12, 119228, Singapore; Khoo Teck Puat - National University Children's Medical Institute, National University Health System, 5 Lower Kent Ridge Road, 119074, Singapore
| | - Tar Choon Aw
- Department of Pathology, Changi General Hospital, 2 Simei Street 3, 529889, Singapore
| | - Soon Kieng Phua
- Department of Pathology, Changi General Hospital, 2 Simei Street 3, 529889, Singapore
| | - Adrian F Low
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, 117597, Singapore; National University Heart Centre, National University Health System, 119074, Singapore
| | - Mark Yan-Yee Chan
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, 117597, Singapore; National University Heart Centre, National University Health System, 119074, Singapore
| | - Gerald F Watts
- School of Medicine, University of Western Australia, 35 Stirling Hwy, Crawley, WA, 6009, Australia; Department of Cardiology, Royal Perth Hospital, Western Australia, Victoria Square, Perth, WA, 6000, Australia
| | - Chew-Kiat Heng
- Department of Paediatrics, Yong Loo Lin School of Medicine, National University of Singapore, 1E Kent Ridge Road, NUHS Tower Block, Level 12, 119228, Singapore; Khoo Teck Puat - National University Children's Medical Institute, National University Health System, 5 Lower Kent Ridge Road, 119074, Singapore
| |
Collapse
|
18
|
Ying Q, Chan DC, Barrett PHR, Watts GF. Unravelling lipoprotein metabolism with stable isotopes: tracing the flow. Metabolism 2021; 124:154887. [PMID: 34508741 DOI: 10.1016/j.metabol.2021.154887] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Revised: 08/16/2021] [Accepted: 09/01/2021] [Indexed: 12/13/2022]
Abstract
Dysregulated lipoprotein metabolism is a major cause of atherosclerotic cardiovascular disease (ASCVD). Use of stable isotope tracers and compartmental modelling have provided deeper understanding of the mechanisms underlying lipid disorders in patients at high risk of ASCVD, including familial hypercholesterolemia (FH), elevated lipoprotein(a) [Lp(a)] and metabolic syndrome (MetS). In patients with FH, deficiency in low-density lipoprotein (LDL) receptor activity not only impairs the catabolism of LDL, but also induces hepatic overproduction and decreases catabolism of triglyceride-rich lipoproteins (TRLs). Patients with elevated Lp(a) are characterized by increased hepatic secretion of Lp(a) particles. Atherogenic dyslipidemia in MetS patients relates to a combination of overproduction of very-low density lipoprotein-apolipoprotein (apo) B-100, decreased catabolism of apoB-100-containing particles, and increased catabolism of high-density lipoprotein-apoA-I particles, as well as to impaired clearance of TRLs in the postprandial state. Kinetic studies show that weight loss, fish oils, statins and fibrates have complementary modes of action that correct atherogenic dyslipidemia. Defining the kinetic mechanisms of action of proprotein convertase subtilisin/kexin type 9 and angiopoietin-like 3 inhibitors on lipid and lipoprotein mechanism in dyslipidemic subjects will further our understanding of these therapies in decreasing the development of ASCVD. "Everything changes but change itself. Everything flows and nothing remains the same... You cannot step twice into the same river, for other waters and yet others go flowing ever on." Heraclitus (c.535- c. 475 BCE).
Collapse
Affiliation(s)
- Qidi Ying
- Medical School, Faculty of Health and Medical Sciences, University of Western Australia, Perth, Australia
| | - Dick C Chan
- Medical School, Faculty of Health and Medical Sciences, University of Western Australia, Perth, Australia
| | - P Hugh R Barrett
- Faculty of Medicine and Health, University of New England, Armidale, Australia
| | - Gerald F Watts
- Medical School, Faculty of Health and Medical Sciences, University of Western Australia, Perth, Australia; Lipid Disorders Clinic, Departments of Cardiology and Internal Medicine, Royal Perth Hospital, Perth, Australia.
| |
Collapse
|
19
|
Longitudinal Assessment of Lipoprotein(a) Levels in Perinatally HIV-Infected Children and Adolescents. Viruses 2021; 13:v13102067. [PMID: 34696496 PMCID: PMC8539147 DOI: 10.3390/v13102067] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Revised: 10/06/2021] [Accepted: 10/12/2021] [Indexed: 11/16/2022] Open
Abstract
HIV is an independent risk factor of cardiovascular disease (CVD); therefore, perinatally HIV-infected (PHIV) children potentially have a greater CVD risk at older age. Lipoprotein(a) (Lp(a)) is an established risk factor for CVD in the general population. To evaluate a potential increased CVD risk for PHIV children, we determined their lipid profiles including Lp(a). In the first substudy, we assessed the lipid profiles of 36 PHIV children visiting the outpatient clinic in Amsterdam between 2012 and 2020. In the second substudy, we enrolled 21 PHIV adolescents and 23 controls matched for age, sex and ethnic background on two occasions with a mean follow-up time of 4.6 years. We assessed trends of lipid profiles and their determinants, including patient and disease characteristics, using mixed models. In the first substudy, the majority of PHIV children were Black (92%) with a median age of 8.0y (5.7–10.8) at first assessment. Persistent elevated Lp(a) levels were present in 21/36 (58%) children (median: 374 mg/L (209–747); cut off = 300). In the second substudy, the median age of PHIV adolescents was 17.5y (15.5–20.7) and of matched controls 16.4y (15.8–19.5) at the second assessment. We found comparable lipid profiles between groups. In both studies, increases in LDL-cholesterol and total cholesterol were associated with higher Lp(a) levels. A majority of PHIV children and adolescents exhibited elevated Lp(a) levels, probably associated with ethnic background. Nonetheless, these elevated Lp(a) levels may additionally contribute to an increased CVD risk.
Collapse
|
20
|
Handhle A, Viljoen A, Wierzbicki AS. Elevated Lipoprotein(a): Background, Current Insights and Future Potential Therapies. Vasc Health Risk Manag 2021; 17:527-542. [PMID: 34526771 PMCID: PMC8436116 DOI: 10.2147/vhrm.s266244] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Accepted: 08/13/2021] [Indexed: 12/13/2022] Open
Abstract
Lipoprotein(a) forms a subfraction of the lipid profile and is characterized by the addition of apolipprotein(a) (apo(a)) to apoB100 derived particles. Its levels are mostly genetically determined inversely related to the number of protein domain (kringle) repeats in apo(a). In epidemiological studies, it shows consistent association with cardiovascular disease (CVD) and most recently with extent of aortic stenosis. Issues with standardizing the measurement of Lp(a) are being resolved and consensus statements favor its measurement in patients at high risk of, or with family histories of CVD events. Major lipid-lowering therapies such as statin, fibrates, and ezetimibe have little effect on Lp(a) levels. Therapies such as niacin or cholesterol ester transfer protein (CETP) inhibitors lower Lp(a) as well as reducing other lipid-related risk factors but have failed to clearly reduce CVD events. Proprotein convertase subtilisin kexin-9 (PCSK9) inhibitors reduce cholesterol and Lp(a) as well as reducing CVD events. New antisense therapies specifically targeting apo(a) and hence Lp(a) have greater and more specific effects and will help clarify the extent to which intervention in Lp(a) levels will reduce CVD events.
Collapse
Affiliation(s)
- Ahmed Handhle
- Department of Metabolic Medicine/Chemical Pathology, Addenbrookes Hospital, Cambridge, UK
| | - Adie Viljoen
- Department of Metabolic Medicine/Chemical Pathology, North & East Hertfordshire Hospitals Trust, Lister Hospital, Hertfordshire, UK
| | - Anthony S Wierzbicki
- Department of Metabolic Medicine/Chemical Pathology, Guy's & St Thomas', Hospitals, London, SE1 7EH, UK
| |
Collapse
|
21
|
Ruscica M, Sirtori CR, Corsini A, Watts GF, Sahebkar A. Lipoprotein(a): Knowns, unknowns and uncertainties. Pharmacol Res 2021; 173:105812. [PMID: 34450317 DOI: 10.1016/j.phrs.2021.105812] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Revised: 08/11/2021] [Accepted: 08/11/2021] [Indexed: 02/07/2023]
Abstract
Over the last 10 years, there have been advances on several aspects of lipoprotein(a) which are reviewed in the present article. Since the standard immunoassays for measuring lipoprotein(a) are not fully apo(a) isoform-insensitive, the application of an LC-MS/MS method for assaying molar concentrations of lipoprotein(a) has been advocated. Genome wide association, epidemiological, and clinical studies have established high lipoprotein(a) as a causal risk factor for atherosclerotic cardiovascular diseases (ASCVD). However, the relative importance of molar concentration, apo(a) isoform size or variants within the LPA gene is still controversial. Lipoprotein(a)-raising single nucleotide polymorphisms has not been shown to add on value in predicting ASCVD beyond lipoprotein(a) concentrations. Although hyperlipoproteinemia(a) represents an important confounder in the diagnosis of familial hypercholesterolemia (FH), it enhances the risk of ASCVD in these patients. Thus, identification of new cases of hyperlipoproteinemia(a) during cascade testing can increase the identification of high-risk individuals. However, it remains unclear whether FH itself increases lipoprotein(a). The ASCVD risk associated with lipoprotein(a) seems to follow a linear gradient across the distribution, regardless of racial subgroups and other risk factors. The inverse association with the risk of developing type 2 diabetes needs consideration as effective lipoprotein(a) lowering therapies are progressing towards the market. Considering that Mendelian randomization analyses have identified the degree of lipoprotein(a)-lowering that is required to achieve ASCVD benefit, the findings of the ongoing outcome trial with pelacarsen will clarify whether dramatically lowering lipoprotein(a) levels can reduce the risk of ASCVD.
Collapse
Affiliation(s)
- Massimiliano Ruscica
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Italy.
| | - Cesare R Sirtori
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Italy
| | - Alberto Corsini
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Italy; IRCCS MultiMedica, Sesto S. Giovanni, Milan, Italy
| | - Gerald F Watts
- School of Medicine, University of Western Australia, Perth, Australia; Lipid Disorders Clinic, Cardiometabolic Services, Department of Cardiology, Royal Perth Hospital, Australia
| | - Amirhossein Sahebkar
- School of Medicine, University of Western Australia, Perth, Australia; Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran; Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
22
|
Marco-Benedí V, Cenarro A, Laclaustra M, Larrea-Sebal A, Jarauta E, Lamiquiz-Moneo I, Calmarza P, Bea AM, Plana N, Pintó X, Martín C, Civeira F. Lipoprotein(a) in hereditary hypercholesterolemia: Influence of the genetic cause, defective gene and type of mutation. Atherosclerosis 2021; 349:211-218. [PMID: 34456049 DOI: 10.1016/j.atherosclerosis.2021.08.009] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Revised: 07/27/2021] [Accepted: 08/05/2021] [Indexed: 02/06/2023]
Abstract
BACKGROUND AND AIMS Lipoprotein(a) [Lp(a)] concentration in heterozygous familial hypercholesterolemia (heFH) is not well established. Whether the genetic defect responsible for heFH plays a role in Lp(a) concentration is unknown. We aimed to compare Lp(a) in controls from a healthy population, in genetically diagnosed heFH and mutation-negative hypercholesterolemia subjects, and to assess the influence on Lp(a) of the genetic defect responsible for heFH. METHODS We conducted a cross-sectional study, performed in a lipid clinic in Spain. We studied adults with suspected heFH and a genetic study of FH genes (LDLR, APOB, APOE and PCSK9) and controls from de Aragon Workers' Health Study. HeFH patients from the Dyslipidemia Registry of the Spanish Atherosclerosis Society (SEA) were used as validation cohort. RESULTS Adjusted geometric means (95% confidence interval) of Lp(a) in controls (n = 1059), heFH (n = 500), and mutation-negative subjects (n = 860) were 14.9 mg/dL (13.6, 16.4), 21.9 mg/dL (18.1, 25.6) and 37.4 mg/dL (33.3, 42.1), p < 0.001 in all comparisons. Among heFH subjects, APOB-dependent FH showed the highest Lp(a), 36.5 mg/dL (22.0, 60.8), followed by LDLR-dependent FH, 21.7 mg/dL (17.9, 26.4). These differences were also observed in heFH from the SEA cohort. The number of plasminogen-like kringle IV type-2 repeats of LPA, the hypercholesterolemia polygenic score or LDLc concentration did not explain these differences. In LDLR-dependent FH, Lp(a) levels were not different depending on the affected protein domain. CONCLUSIONS Lp(a) is elevated in mutation-negative subjects and in heFH. The concentration of Lp(a) in heFH varies in relation to the responsible gene. Higher Lp(a) in heFH is not explained by their higher LDLc.
Collapse
Affiliation(s)
- Victoria Marco-Benedí
- Hospital Universitario Miguel Servet, IIS Aragón, CIBERCV, Zaragoza, Spain; Department of Medicine, Psychiatry and Dermatology, Universidad de Zaragoza, Zaragoza, Spain
| | - Ana Cenarro
- Hospital Universitario Miguel Servet, IIS Aragón, CIBERCV, Zaragoza, Spain
| | - Martín Laclaustra
- Hospital Universitario Miguel Servet, IIS Aragón, CIBERCV, Zaragoza, Spain; Department of Medicine, Psychiatry and Dermatology, Universidad de Zaragoza, Zaragoza, Spain.
| | - Asier Larrea-Sebal
- Fundación Biofisika Bizkaia, Leioa, Spain; Biofisika Institute (UPV/EHU, CSIC), Leioa, Spain, Department of Biochemistry and Molecular Biology, Universidad del País Vasco UPV/EHU, Bilbao, Spain
| | - Estíbaliz Jarauta
- Hospital Universitario Miguel Servet, IIS Aragón, CIBERCV, Zaragoza, Spain; Department of Medicine, Psychiatry and Dermatology, Universidad de Zaragoza, Zaragoza, Spain
| | - Itziar Lamiquiz-Moneo
- Hospital Universitario Miguel Servet, IIS Aragón, CIBERCV, Zaragoza, Spain; Department of Medicine, Psychiatry and Dermatology, Universidad de Zaragoza, Zaragoza, Spain
| | - Pilar Calmarza
- Hospital Universitario Miguel Servet, IIS Aragón, CIBERCV, Zaragoza, Spain
| | - Ana M Bea
- Hospital Universitario Miguel Servet, IIS Aragón, CIBERCV, Zaragoza, Spain
| | - Núria Plana
- Unitat de Medicina Vascular i Metabolisme (UVASMET) Hospital Universitari Sant Joan, IISPV, CIBERDEM, Universitat Rovira i Virgili, Reus, Tarragona, Spain
| | - Xavier Pintó
- Unidad de Lípidos, Servicio de Medicina Interna, Hospital Universitario de Bellvitge-Idibell, Universidad de Barcelona, CiberObn, Barcelona, Spain
| | - César Martín
- Fundación Biofisika Bizkaia, Leioa, Spain; Biofisika Institute (UPV/EHU, CSIC), Leioa, Spain, Department of Biochemistry and Molecular Biology, Universidad del País Vasco UPV/EHU, Bilbao, Spain
| | - Fernando Civeira
- Hospital Universitario Miguel Servet, IIS Aragón, CIBERCV, Zaragoza, Spain; Department of Medicine, Psychiatry and Dermatology, Universidad de Zaragoza, Zaragoza, Spain.
| |
Collapse
|
23
|
Blanchard V, Chemello K, Hollstein T, Hong-Fong CC, Schumann F, Grenkowitz T, Nativel B, Coassin S, Croyal M, Kassner U, Lamina C, Steinhagen-Thiessen E, Lambert G. The size of apolipoprotein (a) is an independent determinant of the reduction in lipoprotein (a) induced by PCSK9 inhibitors. Cardiovasc Res 2021; 118:2103-2111. [PMID: 34314498 DOI: 10.1093/cvr/cvab247] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Accepted: 07/24/2021] [Indexed: 12/14/2022] Open
Abstract
AIMS Lipoprotein (a) [Lp(a)] is a lipoprotein species causatively associated with atherosclerosis. Unlike statins, PCSK9 inhibitors (PCSK9i) reduce Lp(a), but this reduction is highly variable. Levels of Lp(a) are chiefly governed by the size of its signature protein, apolipoprotein (a) [apo(a)]. Whether this parameter determines some of the reduction in Lp(a) induced by PCSK9i remains unknown. We aimed to investigate if the Lp(a) lowering efficacy of PCSK9i is modulated by the size of apo(a), which is genetically determined by the variable number of KIV domains present on that protein. METHODS AND RESULTS The levels of Lp(a) and the size of apo(a) were assessed in plasma samples from 268 patients before and after treatment with PCSK9i. Patients were recruited at the Outpatient Lipid Clinic of the Charité Hospital (Berlin) between 2015 and 2020. They were hypercholesterolemic at very high CVD risk with LDL-cholesterol levels above therapeutic targets despite maximally tolerated lipid-lowering therapy. Patients received either Alirocumab (75 or 150 mg) or Evolocumab (140 mg) every 2 weeks. Apo(a), apoB100, and apoE concentrations as well as apoE major isoforms were determined by liquid chromatography high-resolution mass spectrometry. Apo(a) isoforms sizes were determined by Western Blot. PCSK9i sharply reduced LDL-cholesterol (-57%), apoB100 (-47%) and Lp(a) (-36%). There was a positive correlation between the size of apo(a) and the relative reduction in Lp(a) induced by PCSK9i (r = 0.363, p = 0.0001). The strength of this association remained unaltered after adjustment for baseline Lp(a) levels and all other potential confounding factors. In patients with two detectable apo(a) isoforms, there was also a positive correlation between the size of apo(a) and the reduction in Lp(a), separately for the smaller (r = 0.350, p = 0.0001) and larger (r = 0.324, p = 0.0003) isoforms. The relative contribution of the larger isoform to the total concentration of apo(a) was reduced from 29% to 15% (p < 0.0001). CONCLUSIONS The size of apo(a) is an independent determinant of the response to PCSK9i. Each additional kringle domain is associated with a 3% additional reduction in Lp(a). This explains in part the variable efficacy of PCSK9i and allows to identify patients who will benefit most from these therapies in terms of Lp(a) lowering. TRANSLATIONAL PERSPECTIVE Unlike statins, PCSK9 inhibitors reduce the circulating levels of the highly atherogenic Lipoprotein (a). The underlying mechanism remains a matter of considerable debate. The size of apo(a), the signature protein of Lp(a), is extremely variable (300 to more than 800 kDa) and depends on its number of kringle domains. We now show that each increase in apo(a) size by one kringle domain is associated with a 3% additional reduction in Lp(a) following PCSK9i treatment and that apo(a) size polymorphism is an independent predictor of the reduction in Lp(a) induced by these drugs. In an era of personalized medicine, this allows to identify patients who will benefit most from PCSK9i in terms of Lp(a) lowering.
Collapse
Affiliation(s)
- Valentin Blanchard
- Université de La Réunion, INSERM UMR 1188 DéTROI, Sainte-Clotilde, France.,Centre for Heart & Lung Innovation, St. Paul's Hospital, Vancouver, Canada; Department of Medicine, UBC, Vancouver, Canada
| | - Kévin Chemello
- Université de La Réunion, INSERM UMR 1188 DéTROI, Sainte-Clotilde, France
| | - Tim Hollstein
- Department of Endocrinology, Campus Virchow-Klinikum, Charité Universitätsmedizin, Berlin, Germany.,Division of Endocrinology, Diabetology and Clinical Nutrition, Department of Internal Medicine 1, University of Kiel, Kiel, Germany
| | | | - Friederike Schumann
- Department of Endocrinology, Campus Virchow-Klinikum, Charité Universitätsmedizin, Berlin, Germany
| | - Thomas Grenkowitz
- Department of Endocrinology, Campus Virchow-Klinikum, Charité Universitätsmedizin, Berlin, Germany
| | - Brice Nativel
- Université de La Réunion, INSERM UMR 1188 DéTROI, Sainte-Clotilde, France
| | - Stefan Coassin
- Institute of Genetic Epidemiology, Department of Genetics and Pharmacology, Medical University of Innsbrück, Innsbrück, Austria
| | - Mikaël Croyal
- NUN, INRA, CHU Nantes, UMR 1280, PhAN, IMAD, CRNH-O, Nantes, France
| | - Ursula Kassner
- Department of Endocrinology, Campus Virchow-Klinikum, Charité Universitätsmedizin, Berlin, Germany
| | - Claudia Lamina
- Institute of Genetic Epidemiology, Department of Genetics and Pharmacology, Medical University of Innsbrück, Innsbrück, Austria
| | | | - Gilles Lambert
- Université de La Réunion, INSERM UMR 1188 DéTROI, Sainte-Clotilde, France
| |
Collapse
|
24
|
Khalil YA, Rabès JP, Boileau C, Varret M. APOE gene variants in primary dyslipidemia. Atherosclerosis 2021; 328:11-22. [PMID: 34058468 DOI: 10.1016/j.atherosclerosis.2021.05.007] [Citation(s) in RCA: 64] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Revised: 04/30/2021] [Accepted: 05/12/2021] [Indexed: 01/10/2023]
Abstract
Apolipoprotein E (apoE) is a major apolipoprotein involved in lipoprotein metabolism. It is a polymorphic protein and different isoforms are associated with variations in lipid and lipoprotein levels and thus cardiovascular risk. The isoform apoE4 is associated with an increase in LDL-cholesterol levels and thus a higher cardiovascular risk compared to apoE3. Whereas, apoE2 is associated with a mild decrease in LDL-cholesterol levels. In the presence of other risk factors, apoE2 homozygotes could develop type III hyperlipoproteinemia (familial dysbetalipoproteinemia or FD), an atherogenic disorder characterized by an accumulation of remnants of triglyceride-rich lipoproteins. Several rare APOE gene variants were reported in different types of dyslipidemias including FD, familial combined hyperlipidemia (FCH), lipoprotein glomerulopathy and bona fide autosomal dominant hypercholesterolemia (ADH). ADH is characterized by elevated LDL-cholesterol levels leading to coronary heart disease, and due to molecular alterations in three main genes: LDLR, APOB and PCSK9. The identification of the APOE-p.Leu167del variant as the causative molecular element in two different ADH families, paved the way to considering APOE as a candidate gene for ADH. Due to non mendelian interacting factors, common genetic and environmental factors and perhaps epigenetics, clinical presentation of lipid disorders associated with APOE variants often strongly overlap. More studies are needed to determine the spectrum of APOE implication in each of the diseases, notably ADH, in order to improve clinical and genetic diagnosis, prognosis and patient management. The purpose of this review is to comment on these APOE variants and on the molecular and clinical overlaps between dyslipidemias.
Collapse
Affiliation(s)
- Yara Abou Khalil
- Laboratory for Vascular Translational Science (LVTS), INSERM U1148, Centre Hospitalo-Universitaire Xavier Bichat, Paris, France; Université de Paris, Paris, France; Laboratory of Biochemistry and Molecular Therapeutics (LBTM), Faculty of Pharmacy, Pôle Technologie- Santé (PTS), Saint-Joseph University, Beirut, Lebanon
| | - Jean-Pierre Rabès
- Laboratory for Vascular Translational Science (LVTS), INSERM U1148, Centre Hospitalo-Universitaire Xavier Bichat, Paris, France; Laboratory of Biochemistry and Molecular Genetics, Centre Hospitalo-Universitaire Ambroise Paré, HUPIFO, AP-HP. Paris-Saclay, Boulogne-Billancourt, France; UFR Simone Veil-Santé, UVSQ, Montigny-Le-Bretonneux, France
| | - Catherine Boileau
- Laboratory for Vascular Translational Science (LVTS), INSERM U1148, Centre Hospitalo-Universitaire Xavier Bichat, Paris, France; Université de Paris, Paris, France; Genetics Department, AP-HP, CHU Xavier Bichat, Paris, France
| | - Mathilde Varret
- Laboratory for Vascular Translational Science (LVTS), INSERM U1148, Centre Hospitalo-Universitaire Xavier Bichat, Paris, France; Université de Paris, Paris, France.
| |
Collapse
|
25
|
Ferguson AC, Tank R, Lyall LM, Ward J, Celis-Morales C, Strawbridge R, Ho F, Whelan CD, Gill J, Welsh P, Anderson JJ, Mark PB, Mackay DF, Smith DJ, Pell JP, Cavanagh J, Sattar N, Lyall DM. Alzheimer's Disease Susceptibility Gene Apolipoprotein E (APOE) and Blood Biomarkers in UK Biobank (N = 395,769). J Alzheimers Dis 2021; 76:1541-1551. [PMID: 32651323 DOI: 10.3233/jad-200338] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
BACKGROUND Alzheimer's disease (AD) is a neurodegenerative condition where the underlying etiology is still unclear. Investigating the potential influence of apolipoprotein E (APOE), a major genetic risk factor, on common blood biomarkers could provide a greater understanding of the mechanisms of AD and dementia risk. OBJECTIVE Our objective was to conduct the largest (to date) single-protocol investigation of blood biomarkers in the context of APOE genotype, in UK Biobank. METHODS After quality control and exclusions, data on 395,769 participants of White European ancestry were available for analysis. Linear regressions were used to test potential associations between APOE genotypes and biomarkers. RESULTS Several biomarkers significantly associated with APOEɛ4 'risk' and ɛ2 'protective' genotypes (versus neutral ɛ3/ɛ3). Most associations supported previous data: for example, ɛ4 genotype was associated with elevated low-density lipoprotein cholesterol (LDL) (standardized beta [b] = 0.150 standard deviations [SDs] per allele, p < 0.001) and ɛ2 with lower LDL (b = -0.456 SDs, p < 0.001). There were however instances of associations found in unexpected directions: e.g., ɛ4 and increased insulin-like growth factor (IGF-1) (b = 0.017, p < 0.001) where lower levels have been previously suggested as an AD risk factor. CONCLUSION These findings highlight biomarker differences in non-demented people at genetic risk for dementia. The evidence herein supports previous hypotheses of involvement from cardiometabolic and neuroinflammatory pathways.
Collapse
Affiliation(s)
- Amy C Ferguson
- Institute of Health and Wellbeing, University of Glasgow, Scotland, UK
| | - Rachana Tank
- Institute of Health and Wellbeing, University of Glasgow, Scotland, UK
| | - Laura M Lyall
- Institute of Health and Wellbeing, University of Glasgow, Scotland, UK
| | - Joey Ward
- Institute of Health and Wellbeing, University of Glasgow, Scotland, UK
| | - Carlos Celis-Morales
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, Scotland, UK.,GEEAFyS, Universidad Católica del Maule, Talca, Chile
| | - Rona Strawbridge
- Institute of Health and Wellbeing, University of Glasgow, Scotland, UK.,Health Data Research UK.,Department of Medicine Solna, Cardiovascular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Frederick Ho
- Institute of Health and Wellbeing, University of Glasgow, Scotland, UK
| | | | - Jason Gill
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, Scotland, UK
| | - Paul Welsh
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, Scotland, UK
| | - Jana J Anderson
- Institute of Health and Wellbeing, University of Glasgow, Scotland, UK
| | - Patrick B Mark
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, Scotland, UK
| | - Daniel F Mackay
- Institute of Health and Wellbeing, University of Glasgow, Scotland, UK
| | - Daniel J Smith
- Institute of Health and Wellbeing, University of Glasgow, Scotland, UK
| | - Jill P Pell
- Institute of Health and Wellbeing, University of Glasgow, Scotland, UK
| | - Jonathan Cavanagh
- Institute of Health and Wellbeing, University of Glasgow, Scotland, UK
| | - Naveed Sattar
- Institute of Health and Wellbeing, University of Glasgow, Scotland, UK
| | - Donald M Lyall
- Institute of Health and Wellbeing, University of Glasgow, Scotland, UK
| |
Collapse
|
26
|
Han X, Ong JS, Hewitt AW, Gharahkhani P, MacGregor S. The effects of eight serum lipid biomarkers on age-related macular degeneration risk: a Mendelian randomization study. Int J Epidemiol 2021; 50:325-336. [PMID: 33211829 DOI: 10.1093/ije/dyaa178] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/18/2020] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND Age-related macular degeneration (AMD) is a leading cause of vision loss. Whereas lipids have been studied extensively to understand their effects on cardiovascular diseases, their relationship with AMD remains unclear. METHODS Two-sample Mendelian randomization (MR) analyses were performed to systematically evaluate the causal relationships between eight serum lipid biomarkers, consisting of apolipoprotein A1 (ApoA1), apolipoprotein B (ApoB), total cholesterol (CHOL), high-density lipoprotein cholesterol (HDL-C), direct low-density lipoprotein cholesterol (LDL-C), lipoprotein A [Lp(a)], triglycerides (TG) and non-HDL cholesterol (non-HDL-C), and the risk of different AMD stages and subtypes. We derived 64-407 genetic instruments for eight serum lipid biomarkers in 419 649 participants of European descent from the UK Biobank cohort. We conducted genome-wide association studies (GWAS) for 12 711 advanced AMD cases [8544 choroidal neovascularization (CNV) and 2656 geographic atrophy (GA) specific AMD subtypes] and 5336 intermediate AMD cases with 14 590 controls of European descent from the International AMD Genomics Consortium. RESULTS Higher genetically predicted HDL-C and ApoA1 levels increased the risk of all AMD subtypes. LDL-C, ApoB, CHOL and non-HDL-C levels were associated with decreased risk of intermediate and GA AMD but not with CNV. Genetically predicted TG levels were associated with decreased risk of different AMD subtypes. Sensitivity analyses revealed no evidence for directional pleiotropy effects. In our multivariable MR analyses, adjusting for the effects of correlated lipid biomarkers yielded similar results. CONCLUSION These results suggest the role of lipid metabolism in drusen formation and particularly in AMD development at the early and intermediate stages. Mechanistic studies are warranted to investigate the utility of lipid pathways for therapeutic treatment in preventing AMD.
Collapse
Affiliation(s)
- Xikun Han
- Statistical Genetics, QIMR Berghofer Medical Research Institute, Brisbane, Australia.,School of Medicine, University of Queensland, Brisbane, Australia
| | - Jue-Sheng Ong
- Statistical Genetics, QIMR Berghofer Medical Research Institute, Brisbane, Australia
| | - Alex W Hewitt
- Menzies Institute for Medical Research, University of Tasmania, Australia.,Centre for Eye Research Australia, University of Melbourne, Australia
| | - Puya Gharahkhani
- Statistical Genetics, QIMR Berghofer Medical Research Institute, Brisbane, Australia
| | - Stuart MacGregor
- Statistical Genetics, QIMR Berghofer Medical Research Institute, Brisbane, Australia
| |
Collapse
|
27
|
Abstract
PURPOSE OF REVIEW The functions, genetic variations and impact of apolipoprotein E on lipoprotein metabolism in general are placed in the context of clinical practice dealing with moderate dyslipidaemia as well as dysbetalipoproteinemia, a highly atherogenic disorder and lipoprotein glomerulopathy. RECENT FINDINGS Additional variants of apolipoprotein E and participation of apolipoprotein E in inflammation are of interest. The mostly favourable effects of apolipoprotein E2 as well as the atherogenic nature of apolipoproteinE4, which has an association with cognitive impairment, are confirmed. The contribution of remnant lipoproteins of triglyceride-rich lipoproteins, of which dysbetalipoproteinemia represents an extreme, is explored in atherosclerosis. Mimetic peptides may present new therapeutic approaches. Apolipoprotein E is an important determinant of the lipid profile and cardiovascular health in the population at large and can precipitate dysbetalipoproteinemia and glomerulopathy. Awareness of apolipoprotein E polymorphisms should improve medical care.
Collapse
|
28
|
Hoekstra M, Chen HY, Rong J, Dufresne L, Yao J, Guo X, Tsai MY, Tsimikas S, Post WS, Vasan RS, Rotter JI, Larson MG, Thanassoulis G, Engert JC. Genome-Wide Association Study Highlights APOH as a Novel Locus for Lipoprotein(a) Levels-Brief Report. Arterioscler Thromb Vasc Biol 2021; 41:458-464. [PMID: 33115273 PMCID: PMC7769958 DOI: 10.1161/atvbaha.120.314965] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Accepted: 10/12/2020] [Indexed: 02/07/2023]
Abstract
OBJECTIVE Lp(a) (lipoprotein[a]) is an independent risk factor for cardiovascular diseases and plasma levels are primarily determined by variation at the LPA locus. We performed a genome-wide association study in the UK Biobank to determine whether additional loci influence Lp(a) levels. Approach and Results: We included 293 274 White British individuals in the discovery analysis. Approximately 93 095 623 variants were tested for association with natural log-transformed Lp(a) levels using linear regression models adjusted for age, sex, genotype batch, and 20 principal components of genetic ancestry. After quality control, 131 independent variants were associated at genome-wide significance (P≤5×10-8). In addition to validating previous associations at LPA, APOE, and CETP, we identified a novel variant at the APOH locus, encoding β2GPI (beta2-glycoprotein I). The APOH variant rs8178824 was associated with increased Lp(a) levels (β [95% CI] [ln nmol/L], 0.064 [0.047-0.081]; P=2.8×10-13) and demonstrated a stronger effect after adjustment for variation at the LPA locus (β [95% CI] [ln nmol/L], 0.089 [0.076-0.10]; P=3.8×10-42). This association was replicated in a meta-analysis of 5465 European-ancestry individuals from the Framingham Offspring Study and Multi-Ethnic Study of Atherosclerosis (β [95% CI] [ln mg/dL], 0.16 [0.044-0.28]; P=0.0071). CONCLUSIONS In a large-scale genome-wide association study of Lp(a) levels, we identified APOH as a novel locus for Lp(a) in individuals of European ancestry. Additional studies are needed to determine the precise role of β2GPI in influencing Lp(a) levels as well as its potential as a therapeutic target.
Collapse
Affiliation(s)
- Mary Hoekstra
- Division of Experimental Medicine, McGill University, Montreal, Quebec
- Preventive and Genomic Cardiology, McGill University Health Centre and Research Institute, Montreal, Quebec
| | - Hao Yu Chen
- Division of Experimental Medicine, McGill University, Montreal, Quebec
- Preventive and Genomic Cardiology, McGill University Health Centre and Research Institute, Montreal, Quebec
| | - Jian Rong
- Boston University’s and NHLBI’s Framingham Heart Study, Boston, Massachusetts
| | - Line Dufresne
- Preventive and Genomic Cardiology, McGill University Health Centre and Research Institute, Montreal, Quebec
| | - Jie Yao
- The Institute for Translational Genomics and Population Sciences, Department of Pediatrics, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, California
| | - Xiuqing Guo
- The Institute for Translational Genomics and Population Sciences, Department of Pediatrics, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, California
| | - Michael Y. Tsai
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, Minnesota
| | - Sotirios Tsimikas
- Division of Cardiovascular Medicine, Sulpizio Cardiovascular Center, University of California San Diego, La Jolla, California
| | - Wendy S. Post
- Division of Cardiology, Department of Medicine, The Johns Hopkins University School of Medicine, Baltimore, Maryland
| | | | - Jerome I. Rotter
- The Institute for Translational Genomics and Population Sciences, Department of Pediatrics, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, California
| | - Martin G. Larson
- Boston University’s and NHLBI’s Framingham Heart Study, Boston, Massachusetts
| | - George Thanassoulis
- Division of Experimental Medicine, McGill University, Montreal, Quebec
- Preventive and Genomic Cardiology, McGill University Health Centre and Research Institute, Montreal, Quebec
| | - James C. Engert
- Division of Experimental Medicine, McGill University, Montreal, Quebec
- Preventive and Genomic Cardiology, McGill University Health Centre and Research Institute, Montreal, Quebec
- Department of Human Genetics, McGill University, Montreal, Quebec
| |
Collapse
|
29
|
Willeit P, Yeang C, Moriarty PM, Tschiderer L, Varvel SA, McConnell JP, Tsimikas S. Low-Density Lipoprotein Cholesterol Corrected for Lipoprotein(a) Cholesterol, Risk Thresholds, and Cardiovascular Events. J Am Heart Assoc 2020; 9:e016318. [PMID: 33222611 PMCID: PMC7763787 DOI: 10.1161/jaha.119.016318] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/02/2020] [Accepted: 08/24/2020] [Indexed: 01/18/2023]
Abstract
Background Conventional "low-density lipoprotein cholesterol (LDL-C)" assays measure cholesterol content in both low-density lipoprotein and lipoprotein(a) particles. To clarify the consequences of this methodological limitation for clinical care, our study aimed to compare associations of "LDL-C" and corrected LDL-C with risk of cardiovascular disease and to assess the impact of this correction on the classification of patients into guideline-recommended LDL-C categories. Methods and Results Lipoprotein(a) cholesterol content was estimated as 30% of lipoprotein(a) mass and subtracted from "LDL-C" to obtain corrected LDL-C values (LDL-Ccorr30). Hazard ratios for cardiovascular disease (defined as coronary heart disease, stroke, or coronary revascularization) were quantified by individual-patient-data meta-analysis of 5 statin landmark trials from the Lipoprotein(a) Studies Collaboration (18 043 patients; 5390 events; 4.7 years median follow-up). When comparing top versus bottom quartiles, the multivariable-adjusted hazard ratio for cardiovascular disease was significant for "LDL-C" (1.17; 95% CI, 1.05-1.31; P=0.005) but not for LDL-Ccorr30 (1.07; 95% CI, 0.93-1.22; P=0.362). In a routine laboratory database involving 531 144 patients, reclassification of patients across guideline-recommended LDL-C categories when using LDL-Ccorr30 was assessed. In "LDL-C" categories of 70 to <100, 100 to <130, 130 to <190, and ≥190 mg/dL, significant proportions (95% CI) of participants were reassigned to lower LDL-C categories when LDL-Ccorr30 was used: 30.2% (30.0%-30.4%), 35.1% (34.9%-35.4%), 32.9% (32.6%-33.1%), and 41.1% (40.0%-42.2%), respectively. Conclusions "LDL-C" was associated with incident cardiovascular disease only when lipoprotein(a) cholesterol content was included in its measurement. Refinement in techniques to accurately measure LDL-C, particularly in patients with elevated lipoprotein(a) levels, is warranted to assign risk to the responsible lipoproteins.
Collapse
Affiliation(s)
- Peter Willeit
- Department of NeurologyMedical University of InnsbruckInnsbruckAustria
- Department of Public Health and Primary CareUniversity of CambridgeCambridgeUK
| | - Calvin Yeang
- Division of Cardiovascular MedicineSulpizio Cardiovascular CenterUniversity of California, San DiegoLa JollaCA
| | - Patrick M. Moriarty
- Division of Clinical PharmacologyDepartment of Internal MedicineUniversity of Kansas Medical CenterKansas CityMO
| | - Lena Tschiderer
- Department of NeurologyMedical University of InnsbruckInnsbruckAustria
| | | | | | - Sotirios Tsimikas
- Division of Cardiovascular MedicineSulpizio Cardiovascular CenterUniversity of California, San DiegoLa JollaCA
| |
Collapse
|
30
|
Gharbavi M, Johari B, Eslami SS, Mousazadeh N, Sharafi A. Cholesterol-conjugated bovine serum albumin nanoparticles as a tamoxifen tumor-targeted delivery system. Cell Biol Int 2020; 44:2485-2498. [PMID: 32841441 DOI: 10.1002/cbin.11455] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Revised: 08/18/2020] [Accepted: 08/23/2020] [Indexed: 12/21/2022]
Abstract
In the present study, we introduced cholesterol (CLO)-conjugated bovine serum albumin nanoparticles (BSA NPs) as a new system for indirect targeting drug delivery. Tamoxifen, as an anticancer drug, was loaded on BSA NPs (BSA-TAX NPs); CLO was then conjugated to the BSA-TAX NPs surface for the targeted delivery of NPs system, by 1-ethyl-3-(3-dimethylaminopropyl) carbodiimide/N-hydroxy succinimide carbodiimide chemistry (CLO-BSA-TAX NPs). The physicochemical properties, toxicity, in vitro, and in vivo biocompatibility of the BSA NPs system were characterized on cancer cell lines (4T1). The results revealed that the BSA NPs system has a regular spherical shape and negative zeta-potential values. The drug release of BSA NPs system has shown controlled and pH-dependent drug release behavior. BSA NPs system was biocompatible but it was potentially toxic on the cancer cell line. The CLO-BSA-TAX NPs exhibited higher toxicity against cancer cell lines than other NPs formulation (BSA NPs and BSA-TAX NPs). It can be concluded that the CLO, as an indirect targeting agent, enhances the toxicity and specificity of NPs system on cancer cell lines. It could potentially be suitable approaches to targeting the tumors in clinical cancer therapy.
Collapse
Affiliation(s)
- Mahmoud Gharbavi
- Student Research Committee, Zanjan University of Medical Sciences, Zanjan, Iran.,Department of Pharmaceutical Biomaterials, School of Pharmacy, Zanjan University of Medical Sciences, Zanjan, Iran.,Zanjan Pharmaceutical Biotechnology Research Center, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Behrooz Johari
- Cancer Gene Therapy Research Center, Zanjan University of Medical Sciences, Zanjan, Iran.,Department of Medical Biotechnology, School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Seyed Sadegh Eslami
- Department of Medical Biotechnology, Faculty of Allied Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Navid Mousazadeh
- Department of Medical Biotechnology, School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Ali Sharafi
- Zanjan Pharmaceutical Biotechnology Research Center, Zanjan University of Medical Sciences, Zanjan, Iran
| |
Collapse
|
31
|
Moriarty PM, Gorby LK, Stroes ES, Kastelein JP, Davidson M, Tsimikas S. Lipoprotein(a) and Its Potential Association with Thrombosis and Inflammation in COVID-19: a Testable Hypothesis. Curr Atheroscler Rep 2020; 22:48. [PMID: 32710255 PMCID: PMC7381416 DOI: 10.1007/s11883-020-00867-3] [Citation(s) in RCA: 60] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
PURPOSE OF REVIEW The COVID-19 pandemic has infected over > 11 million as of today people worldwide and is associated with significant cardiovascular manifestations, particularly in subjects with preexisting comorbidities and cardiovascular risk factors. Recently, a predisposition for arterial and venous thromboses has been reported in COVID-19 infection. We hypothesize that besides conventional risk factors, subjects with elevated lipoprotein(a) (Lp(a)) may have a particularly high risk of developing cardiovascular complications. RECENT FINDINGS The Lp(a) molecule has the propensity for inhibiting endogenous fibrinolysis through its apolipoprotein(a) component and for enhancing proinflammatory effects such as through its content of oxidized phospholipids. The LPA gene contains an interleukin-6 (IL-6) response element that may induce an acute phase-type increase in Lp(a) levels following a cytokine storm from COVID-19. Thus, subjects with either baseline elevated Lp(a) or those who have an increase following COVID-19 infection, or both, may be at very high risk of developing thromboses. Elevated Lp(a) may also lead to acute destabilization of preexisting but quiescent atherosclerotic plaques, which might induce acute myocardial infarction and stroke. Ongoing studies with IL-6 antagonists may be informative in understanding this relationship, and registries are being initiated to measure Lp(a) in subjects infected with COVID-19. If indeed an association is suggestive of being causal, consideration can be given to systematic testing of Lp(a) and prophylactic systemic anticoagulation in infected inpatients. Therapeutic lipid apheresis and pharmacotherapy for the reduction of Lp(a) levels may minimize thrombogenic potential and proinflammatory effects. We propose studies to test the hypothesis that Lp(a) may contribute to cardiovascular complications of COVID-19.
Collapse
Affiliation(s)
- Patrick M Moriarty
- Division of Clinical Pharmacology, University of Kansas Medical Center, Kansas City, KS, USA.
| | - Lauryn K Gorby
- Division of Clinical Pharmacology, University of Kansas Medical Center, Kansas City, KS, USA
| | - Erik S Stroes
- Department of Vascular Medicine, Academic Medical Center, Amsterdam, The Netherlands
| | - John P Kastelein
- Department of Vascular Medicine, Academic Medical Center, Amsterdam, The Netherlands
| | - Michael Davidson
- Lipid Clinic, The University of Chicago Pritzker School of Medicine, Chicago, IL, USA
| | - Sotirios Tsimikas
- Division of Cardiovascular Medicine, University of California San Diego, La Jolla, CA, USA.
- Vascular Medicine Program, Sulpizio Cardiovascular Center, University of California San Diego, 9500 Gilman Drive, BSB 1080, La Jolla, CA, 92093-0682, USA.
| |
Collapse
|
32
|
Korostovtseva L, Alieva A, Rotar O, Bochkarev M, Boyarinova M, Sviryaev Y, Konradi A, Shlyakhto E. Sleep Duration, Lipid Profile and Insulin Resistance: Potential Role of Lipoprotein(a). Int J Mol Sci 2020; 21:E4680. [PMID: 32630105 PMCID: PMC7369827 DOI: 10.3390/ijms21134680] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Revised: 06/22/2020] [Accepted: 06/28/2020] [Indexed: 11/29/2022] Open
Abstract
Lipoprotein (a) (Lp(a)) is considered a genetic factor for cardiovascular disease playing an important role in atherogenesis and thrombosis, but the evidence about its association with sleep duration is controversial. We evaluated the relation between self-reported sleep duration and Lp(a). Among 1600 participants of the population-based sample, we selected 1427 subjects without previously known cardiovascular events, who answered the questions about their sleep duration; had valid lipid profile results (total cholesterol, low- and high-density lipoproteins, Lp(a), apolipoprotein AI (ApoAI), ApoB, and ApoB/ApoAI); and did not take lipid-lowering drugs (mean age 46 ± 12 years). We performed a structured interview, which included questions about lifestyle, medical history, complaints, and sleep duration (How long have you been sleeping per night during the last month?). Sleep duration was classified as follows: <6 h/night-short, 6-9 h/night-normal, and ≥10 h/night-long. Overall, 73 respondents (5.2%) were short-sleepers and 69 (4.8%) long-sleepers. Males were slightly more prevalent among short-sleepers. The groups matched by age, body mass index, blood pressure, diabetes mellitus, and hypertension rate. Short-sleepers had lower rates of high total cholesterol (≥5.0 mmol/L), lower Lp(a) levels and lower rates of increased Lp(a) ≥0.5 g/L, and higher insulin and insulin resistance (assessed by the homeostatic model assessment for insulin resistance (HOMA-IR)). ApoAI, ApoB, their ratio, and other lab tests were similar in the groups. The multinomial logistic regression demonstrated that only the short sleep duration was independently (odds ratio (OR) 0.29, 95% confidence interval (CI) (0.09-0.91), p = 0.033) associated with Lp(a) (χ2 = 41.58, p = 0.003). Other influencing factors were smoking and HOMA-IR. Such an association was not found for long-sleepers. In conclusion, a short-sleep duration is associated with Lp(a). The latter might mediate the higher insulin resistance and higher cardiometabolic risks in short-sleepers.
Collapse
Affiliation(s)
- Lyudmila Korostovtseva
- Almazov National Medical Research Centre, St Petersburg 197341, Russia; (A.A.); (O.R.); (M.B.); (M.B.); (Y.S.); (A.K.); (E.S.)
| | - Asiiat Alieva
- Almazov National Medical Research Centre, St Petersburg 197341, Russia; (A.A.); (O.R.); (M.B.); (M.B.); (Y.S.); (A.K.); (E.S.)
| | - Oxana Rotar
- Almazov National Medical Research Centre, St Petersburg 197341, Russia; (A.A.); (O.R.); (M.B.); (M.B.); (Y.S.); (A.K.); (E.S.)
| | - Mikhail Bochkarev
- Almazov National Medical Research Centre, St Petersburg 197341, Russia; (A.A.); (O.R.); (M.B.); (M.B.); (Y.S.); (A.K.); (E.S.)
| | - Maria Boyarinova
- Almazov National Medical Research Centre, St Petersburg 197341, Russia; (A.A.); (O.R.); (M.B.); (M.B.); (Y.S.); (A.K.); (E.S.)
| | - Yurii Sviryaev
- Almazov National Medical Research Centre, St Petersburg 197341, Russia; (A.A.); (O.R.); (M.B.); (M.B.); (Y.S.); (A.K.); (E.S.)
- Sechenov Institute of Evolutionary Physiology and Biochemistry of RAS, St Petersburg 194223, Russia
| | - Aleksandra Konradi
- Almazov National Medical Research Centre, St Petersburg 197341, Russia; (A.A.); (O.R.); (M.B.); (M.B.); (Y.S.); (A.K.); (E.S.)
- Institute of Translational Medicine, ITMO University, St Petersburg 197101, Russia
| | - Eugene Shlyakhto
- Almazov National Medical Research Centre, St Petersburg 197341, Russia; (A.A.); (O.R.); (M.B.); (M.B.); (Y.S.); (A.K.); (E.S.)
| |
Collapse
|
33
|
Affiliation(s)
- Dick C Chan
- Metabolic Research Centre, School of Medicine, Faculty of Medicine and Health Sciences, University of Western Australia, Perth, Western Australia, Australia
| | | |
Collapse
|
34
|
Coassin S, Hermann-Kleiter N, Haun M, Wahl S, Wilson R, Paulweber B, Kunze S, Meitinger T, Strauch K, Peters A, Waldenberger M, Kronenberg F, Lamina C. A genome-wide analysis of DNA methylation identifies a novel association signal for Lp(a) concentrations in the LPA promoter. PLoS One 2020; 15:e0232073. [PMID: 32343731 PMCID: PMC7188291 DOI: 10.1371/journal.pone.0232073] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Accepted: 03/19/2020] [Indexed: 12/24/2022] Open
Abstract
Lipoprotein(a) [Lp(a)] is a major cardiovascular risk factor, which is largely genetically determined by one major gene locus, the LPA gene. Many aspects of the transcriptional regulation of LPA are poorly understood and the role of epigenetics has not been addressed yet. Therefore, we conducted an epigenome-wide analysis of DNA methylation on Lp(a) levels in two population-based studies (total n = 2208). We identified a CpG site in the LPA promoter which was significantly associated with Lp(a) concentrations. Surprisingly, the identified CpG site was found to overlap the SNP rs76735376. We genotyped this SNP de-novo in three studies (total n = 7512). The minor allele of rs76735376 (1.1% minor allele frequency) was associated with increased Lp(a) values (p = 1.01e-59) and explained 3.5% of the variation of Lp(a). Statistical mediation analysis showed that the effect on Lp(a) is rather originating from the base change itself and is not mediated by DNA methylation levels. This finding is supported by eQTL data from 208 liver tissue samples from the GTEx project, which shows a significant association of the rs76735376 minor allele with increased LPA expression. To evaluate, whether the association signal at rs76735376 may actually be derived from a stronger eQTL signal in LD with this SNP, eQTL association results of all correlated SNPs (r2≥0.1) were integrated with genetic association results. This analysis pinpointed to rs10455872 as the potential trigger of the effect of rs76735376. Furthermore, both SNPs coincide with short apo(a) isoforms. Adjusting for both, rs10455872 and the apo(a) isoforms diminished the effect size of rs76735376 to 5.38 mg/dL (p = 0.0463). This indicates that the effect of rs76735376 can be explained by both an independent effect of the SNP and a strong correlation with rs10455872 and apo(a) isoforms.
Collapse
Affiliation(s)
- Stefan Coassin
- Department of Genetics and Pharmacology, Institute of Genetic Epidemiology, Medical University of Innsbruck, Innsbruck, Austria
| | - Natascha Hermann-Kleiter
- Department of Genetics and Pharmacology, Institute of Cell Genetics, Medical University of Innsbruck, Innsbruck, Austria
| | - Margot Haun
- Department of Genetics and Pharmacology, Institute of Genetic Epidemiology, Medical University of Innsbruck, Innsbruck, Austria
| | - Simone Wahl
- Research Unit of Molecular Epidemiology, Helmholtz Zentrum München–German Research Center for Environmental Health, Neuherberg, Germany
- Institute of Epidemiology II, Helmholtz Zentrum München–German Research Center for Environmental Health, Neuherberg, Germany
| | - Rory Wilson
- Research Unit of Molecular Epidemiology, Helmholtz Zentrum München–German Research Center for Environmental Health, Neuherberg, Germany
- Institute of Epidemiology II, Helmholtz Zentrum München–German Research Center for Environmental Health, Neuherberg, Germany
| | - Bernhard Paulweber
- First Department of Internal Medicine, Paracelsus Private Medical University, Salzburg, Austria
| | - Sonja Kunze
- Research Unit of Molecular Epidemiology, Helmholtz Zentrum München–German Research Center for Environmental Health, Neuherberg, Germany
- Institute of Epidemiology II, Helmholtz Zentrum München–German Research Center for Environmental Health, Neuherberg, Germany
| | - Thomas Meitinger
- German Center for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance, Munich, Germany
- Institute of Human Genetics, Technische Universität München, Munich, Germany
- German Research Center for Environmental Health, Institute of Human Genetics, Helmholtz Zentrum München, Neuherberg, Germany
| | - Konstantin Strauch
- German Research Center for Environmental Health, Institute of Genetic Epidemiology, Helmholtz Zentrum München, Neuherberg, Germany
- Institute of Medical Informatics, Biometry, and Epidemiology, Ludwig-Maximilians-Universität, Munich, Germany
- Institute of Medical Biostatistics, Epidemiology and Informatics (IMBEI), University Medical Center, Johannes Gutenberg University, Mainz, Germany
| | - Annette Peters
- Institute of Epidemiology II, Helmholtz Zentrum München–German Research Center for Environmental Health, Neuherberg, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance, Munich, Germany
| | - Melanie Waldenberger
- Research Unit of Molecular Epidemiology, Helmholtz Zentrum München–German Research Center for Environmental Health, Neuherberg, Germany
- Institute of Epidemiology II, Helmholtz Zentrum München–German Research Center for Environmental Health, Neuherberg, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance, Munich, Germany
| | - Florian Kronenberg
- Department of Genetics and Pharmacology, Institute of Genetic Epidemiology, Medical University of Innsbruck, Innsbruck, Austria
| | - Claudia Lamina
- Department of Genetics and Pharmacology, Institute of Genetic Epidemiology, Medical University of Innsbruck, Innsbruck, Austria
| |
Collapse
|
35
|
Shapiro MD, Minnier J, Tavori H, Kassahun H, Flower A, Somaratne R, Fazio S. Relationship Between Low-Density Lipoprotein Cholesterol and Lipoprotein(a) Lowering in Response to PCSK9 Inhibition With Evolocumab. J Am Heart Assoc 2020; 8:e010932. [PMID: 30755061 PMCID: PMC6405654 DOI: 10.1161/jaha.118.010932] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Background Beyond their potent LDL (low‐density lipoprotein) cholesterol (LDL‐C)–lowering efficacy (50–60%), PCSK9 (proprotein convertase subtilisin/kexin type 9) inhibitors also reduce Lp(a) (lipoprotein[a]) levels by 25% to 30%, suggesting a 2:1 response ratio. We aimed to characterize the relationship between LDL‐C and Lp(a) lowering by evolocumab, a PCSK9 inhibitor, in a large clinical trial population and to determine the prevalence of concordant/discordant LDL‐C and Lp(a) responses to PCSK9 inhibition. Methods and Results Data were analyzed from 4 randomized, 12‐week, multicenter, phase 3 evolocumab trials. Patients with familial hypercholesterolemia, nonfamilial hypercholesterolemia, or statin intolerance participated in the trials. The main measure was the degree of concordance or discordance of LDL‐C and Lp(a) in response to PCSK9 inhibition; concordant response was defined as LDL‐C reduction >35% and Lp(a) reduction >10%. The study cohort comprised 895 patients (438 female; median age: 59.0 years [interquartile range: 51–66 years]). Baseline mean level of LDL‐C was 133.6 mg/dL (SE: 1.7) and median Lp(a) level was 46.4 mg/dL (interquartile range: 18.4–82.4 mg/dL). A discordant response was observed in 165 (19.7%) patients. With these cutoffs, the prevalence of discordance was higher when considering baseline Lp(a) concentrations >30 mg/dL (26.5%) or >50 mg/dL (28.6%). Conclusions We demonstrate high prevalence of discordance in LDL‐C and Lp(a) reduction in response to evolocumab, particularly when considering higher baseline Lp(a) concentrations, indicating the possibility of alternative pathways beyond LDLR (LDL receptor)–mediated clearance involved in Lp(a) reduction by evolocumab. Clinical Trial Registration URL: http://www.clinicaltrials.gov. Unique identifiers: NCT01763827, NCT01763866, NCT01763905, NCT01763918. See Editorial by Nestel
Collapse
Affiliation(s)
- Michael D. Shapiro
- Knight Cardiovascular InstituteCenter for Preventive CardiologyOregon Health & Science UniversityPortlandOR
| | - Jessica Minnier
- Knight Cardiovascular InstituteCenter for Preventive CardiologyOregon Health & Science UniversityPortlandOR
- OHSU‐PSU School of Public HealthOregon Health & Science UniversityPortlandOR
| | - Hagai Tavori
- Knight Cardiovascular InstituteCenter for Preventive CardiologyOregon Health & Science UniversityPortlandOR
| | | | | | | | - Sergio Fazio
- Knight Cardiovascular InstituteCenter for Preventive CardiologyOregon Health & Science UniversityPortlandOR
| |
Collapse
|
36
|
Croyal M, Blanchard V, Ouguerram K, Chétiveaux M, Cabioch L, Moyon T, Billon-Crossouard S, Aguesse A, Bernardeau K, Le May C, Flet L, Lambert G, Hadjadj S, Cariou B, Krempf M, Nobécourt-Dupuy E. VLDL (Very-Low-Density Lipoprotein)-Apo E (Apolipoprotein E) May Influence Lp(a) (Lipoprotein [a]) Synthesis or Assembly. Arterioscler Thromb Vasc Biol 2020; 40:819-829. [DOI: 10.1161/atvbaha.119.313877] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Objective:
To clarify the association between PCSK9 (proprotein convertase subtilisin/kexin type 9) and Lp(a) (lipoprotein [a]), we studied Lp(a) kinetics in patients with loss-of-function and gain-of-function
PCSK9
mutations and in patients in whom extended-release niacin reduced Lp(a) and PCSK9 concentrations.
Approach and Results:
Six healthy controls, 9 heterozygous patients with familial hypercholesterolemia (5 with low-density lipoprotein receptor [
LDLR
] mutations and 4 with
PCSK9
gain-of-function mutations) and 3 patients with heterozygous dominant-negative
PCSK9
loss-of-function mutations were included in the preliminary study. Eight patients were enrolled in a second study assessing the effects of 2 g/day extended-release niacin. Apolipoprotein kinetics in VLDL (very-low-density lipoprotein), LDL (low-density lipoprotein), and Lp(a) were studied using stable isotope techniques. Plasma Lp(a) concentrations were increased in
PCSK9
-gain-of-function and familial hypercholesterolemia-
LDLR
groups compared with controls and
PCSK9
-loss-of-function groups (14±12 versus 5±4 mg/dL;
P
=0.04), but no change was observed in Lp(a) fractional catabolic rate. Subjects with
PCSK9
-loss-of-function mutations displayed reduced apoE (apolipoprotein E) concentrations associated with a VLDL-apoE absolute production rate reduction. Lp(a) and VLDL-apoE absolute production rates were correlated (
r
=0.50;
P
<0.05). ApoE-to-apolipoprotein (a) molar ratios in Lp(a) increased with plasma Lp(a) (
r
=0.96;
P
<0.001) but not with PCSK9 levels. Extended-release niacin-induced reductions in Lp(a) and VLDL-apoE absolute production rate were correlated (
r
=0.83;
P
=0.015). In contrast, PCSK9 reduction (−35%;
P
=0.008) was only correlated with that of VLDL-apoE absolute production rate (
r
=0.79;
P
=0.028).
Conclusions:
VLDL-apoE production could determine Lp(a) production and/or assembly. As PCSK9 inhibitors reduce plasma apoE and Lp(a) concentrations, apoE could be the link between PCSK9 and Lp(a).
Collapse
Affiliation(s)
- Mikaël Croyal
- From the NUN, INRA, CHU Nantes, UMR 1280, PhAN, IMAD, CRNH-O, France (M. Croyal, K.O., S.B.-C., A.A., M.K.)
- CRNH-O Mass Spectrometry Core Facility, F-44000 Nantes, France (M. Croyal, K.O., T.M., S.B.-C., A.A., M.K.)
| | - Valentin Blanchard
- Université de La Réunion, INSERM, UMR 1188 Diabète athérothrombose Réunion Océan Indien (DéTROI), Plateforme CYROI, Saint-Denis de La Réunion, France (V.B., G.L.)
| | - Khadija Ouguerram
- From the NUN, INRA, CHU Nantes, UMR 1280, PhAN, IMAD, CRNH-O, France (M. Croyal, K.O., S.B.-C., A.A., M.K.)
- CRNH-O Mass Spectrometry Core Facility, F-44000 Nantes, France (M. Croyal, K.O., T.M., S.B.-C., A.A., M.K.)
| | - Maud Chétiveaux
- L’institut du thorax, INSERM, CNRS, University of Nantes, France (M. Chétiveaux, C.L.M.)
| | - Léa Cabioch
- Biogenouest-Corsaire platform, Saint Gilles, France (L.C.)
| | - Thomas Moyon
- CRNH-O Mass Spectrometry Core Facility, F-44000 Nantes, France (M. Croyal, K.O., T.M., S.B.-C., A.A., M.K.)
| | - Stéphanie Billon-Crossouard
- From the NUN, INRA, CHU Nantes, UMR 1280, PhAN, IMAD, CRNH-O, France (M. Croyal, K.O., S.B.-C., A.A., M.K.)
- CRNH-O Mass Spectrometry Core Facility, F-44000 Nantes, France (M. Croyal, K.O., T.M., S.B.-C., A.A., M.K.)
| | - Audrey Aguesse
- From the NUN, INRA, CHU Nantes, UMR 1280, PhAN, IMAD, CRNH-O, France (M. Croyal, K.O., S.B.-C., A.A., M.K.)
- CRNH-O Mass Spectrometry Core Facility, F-44000 Nantes, France (M. Croyal, K.O., T.M., S.B.-C., A.A., M.K.)
| | - Karine Bernardeau
- P2R «Production de protéines recombinantes», CRCINA, SFR-Santé, INSERM, CNRS, UNIV Nantes, CHU Nantes, France (K.B.)
| | - Cédric Le May
- L’institut du thorax, INSERM, CNRS, University of Nantes, France (M. Chétiveaux, C.L.M.)
| | - Laurent Flet
- Pharmacy Department, Nantes University Hospital, France (L.F.)
| | - Gilles Lambert
- Université de La Réunion, INSERM, UMR 1188 Diabète athérothrombose Réunion Océan Indien (DéTROI), Plateforme CYROI, Saint-Denis de La Réunion, France (V.B., G.L.)
| | - Samy Hadjadj
- L’institut du thorax, INSERM, CNRS, University of Nantes, CHU Nantes, France (S.H., B.C.)
| | - Bertrand Cariou
- L’institut du thorax, INSERM, CNRS, University of Nantes, CHU Nantes, France (S.H., B.C.)
| | - Michel Krempf
- From the NUN, INRA, CHU Nantes, UMR 1280, PhAN, IMAD, CRNH-O, France (M. Croyal, K.O., S.B.-C., A.A., M.K.)
- CRNH-O Mass Spectrometry Core Facility, F-44000 Nantes, France (M. Croyal, K.O., T.M., S.B.-C., A.A., M.K.)
- ELSAN, clinique Bretéché, Nantes, France (M.K.)
| | | |
Collapse
|
37
|
Schatz U, Fischer S, Müller G, Tselmin S, Birkenfeld AL, Julius U, März W, Bornstein SR. Cardiovascular risk factors in patients with premature cardiovascular events attending the University of Dresden Lipid Clinic. ATHEROSCLEROSIS SUPP 2019; 40:94-99. [PMID: 31818455 DOI: 10.1016/j.atherosclerosissup.2019.08.044] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
OBJECTIVES Despite improved treatment, premature cardiovascular (CV) events remain a major health problem. Aim of this study was to evaluate the patterns of risk factors in patients with premature CV events. METHODS CV risk factors (CVRF) were evaluated in 130 patients with a history of CV events (myocardial infarction, stroke, limb ischemia, stent and bypass intervention in any vessel bed) under 50 years of age attending our lipid clinic. Patients were also stratified according to their Lp(a) concentrations: group 1: 0-45 nmol/l (<18 mg/dl); group 2: >45-120 nmol/l (>18-50 mg/dl); group 3: >120 nmol/l (>50 mg/dl). RESULTS The most common risk factors in our patients were male sex (75%), current (61%) and previous smoking (9%), arterial hypertension (70%), and a positive family history of early CV events (54%) and hyperlipidemia (69%). Only 27% had a BMI >30 kg/m2 and 14% had diabetes mellitus. 69% of patients with premature CV disease (CVD) showed Lp(a) levels > 120 nmol/l (>50 mg/dl). Patients with the highest Lp(a) showed a tendency of more frequent positive family histories of hyperlipidemia. They had experienced their first CV event on average 3 years earlier than those with low Lp(a). CV events predominantly involved coronary arteries. 85% of patients experienced at least one coronary event. CONCLUSION In patients with premature CV disease male sex, smoking, hypertension, a positive family history and elevated Lp(a) are the most important CV risk factors. Lp(a) should be considered in the management of young patients with CV disease.
Collapse
Affiliation(s)
- Ulrike Schatz
- Department of Internal Medicine III, Faculty of Medicine Carl Gustav Carus at the Technische Universität Dresden, Fetscherstr. 74, 01307, Dresden, Germany.
| | - Sabine Fischer
- Department of Internal Medicine III, Faculty of Medicine Carl Gustav Carus at the Technische Universität Dresden, Fetscherstr. 74, 01307, Dresden, Germany.
| | - Gabriele Müller
- Center for Evidence-Based Healthcare, Faculty of Medicine Carl Gustav Carus at the Technische Universität Dresden, Fetscherstr. 74, 01307, Dresden, Germany.
| | - Sergey Tselmin
- Department of Internal Medicine III, Faculty of Medicine Carl Gustav Carus at the Technische Universität Dresden, Fetscherstr. 74, 01307, Dresden, Germany.
| | - Andreas L Birkenfeld
- Department of Internal Medicine III, Faculty of Medicine Carl Gustav Carus at the Technische Universität Dresden, Fetscherstr. 74, 01307, Dresden, Germany.
| | - Ulrich Julius
- Department of Internal Medicine III, Faculty of Medicine Carl Gustav Carus at the Technische Universität Dresden, Fetscherstr. 74, 01307, Dresden, Germany.
| | - Winfried März
- Medical Clinic V (Nephrology, Rheumatology, Hypertensiology, Endocrinolgy, Diabetology), Medical Faculty Mannheim, University of Heidelberg, Theodor-Kutzer-Ufer 1-3, 68167, Mannheim, Germany; Clinical Institute of Medical and Chemical Laboratory Diagnostics, Medical University of Graz, Auenbruggerplatz 2, 8036, Graz, Austria; SYNLAB Academy, SYNLAB Holding Deutschland GmbH, P5,7, 68161, Mannheim, Germany.
| | - Stefan R Bornstein
- Department of Internal Medicine III, Faculty of Medicine Carl Gustav Carus at the Technische Universität Dresden, Fetscherstr. 74, 01307, Dresden, Germany.
| |
Collapse
|
38
|
Ward NC, Kostner KM, Sullivan DR, Nestel P, Watts GF. Molecular, Population, and Clinical Aspects of Lipoprotein(a): A Bridge Too Far? J Clin Med 2019; 8:E2073. [PMID: 31783529 PMCID: PMC6947201 DOI: 10.3390/jcm8122073] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2019] [Revised: 11/15/2019] [Accepted: 11/15/2019] [Indexed: 12/16/2022] Open
Abstract
There is now significant evidence to support an independent causal role for lipoprotein(a) (Lp(a)) as a risk factor for atherosclerotic cardiovascular disease. Plasma Lp(a) concentrations are predominantly determined by genetic factors. However, research into Lp(a) has been hampered by incomplete understanding of its metabolism and proatherogeneic properties and by a lack of suitable animal models. Furthermore, a lack of standardized assays to measure Lp(a) and no universal consensus on optimal plasma levels remain significant obstacles. In addition, there are currently no approved specific therapies that target and lower elevated plasma Lp(a), although there are recent but limited clinical outcome data suggesting benefits of such reduction. Despite this, international guidelines now recognize elevated Lp(a) as a risk enhancing factor for risk reclassification. This review summarises the current literature on Lp(a), including its discovery and recognition as an atherosclerotic cardiovascular disease risk factor, attempts to standardise analytical measurement, interpopulation studies, and emerging therapies for lowering elevated Lp(a) levels.
Collapse
Affiliation(s)
- Natalie C. Ward
- School of Public Health, Curtin University, Perth 6102, Australia;
- School of Medicine, University of Western Australia, Perth 6009, Australia
| | - Karam M. Kostner
- Department of Cardiology, Mater Hospital, Brisbane 4104, Australia;
- School of Medicine University of Queensland, Brisbane 4072, Australia
| | - David R. Sullivan
- Medical School, The University of Sydney, Sydney 2006, Australia;
- Charles Perkins Centre, The University of Sydney, Sydney 2006, Australia
- Department of Biochemistry, Royal Prince Alfred Hospital, Sydney 2050, Australia
| | - Paul Nestel
- Baker Heart & Diabetes Institute, Melbourne 3004, Australia;
- Department of Cardiology, The Alfred Hospital, Melbourne 3004, Australia
| | - Gerald F. Watts
- School of Medicine, University of Western Australia, Perth 6009, Australia
- Lipid Disorders Clinic, Department of Cardiology, Royal Perth Hospital, Perth 6000, Australia
| |
Collapse
|
39
|
Van den Hof M, Klein Haneveld MJ, Blokhuis C, Scherpbier HJ, Jansen HPG, Kootstra NA, Dallinga-Thie GM, Van Deventer SJH, Tsimikas S, Pajkrt D. Elevated Lipoprotein(a) in Perinatally HIV-Infected Children Compared With Healthy Ethnicity-Matched Controls. Open Forum Infect Dis 2019; 6:ofz301. [PMID: 31660394 PMCID: PMC6736182 DOI: 10.1093/ofid/ofz301] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2019] [Indexed: 01/06/2023] Open
Abstract
Background HIV-associated cardiovascular disease (CVD) risk in combination antiretroviral therapy (cART)-treated perinatally HIV-infected patients (PHIV+) remains unknown due to the young age of this population. Lipoprotein(a) (Lp(a)) has been established as an independent causal risk factor for CVD in the general population but has not been well established in the population of PHIV+. Methods We cross-sectionally compared lipid profiles, including nonfasting Lp(a), together with total cholesterol, high-density lipoprotein cholesterol, low-density lipoprotein cholesterol, and triglycerides between 35 cART-treated PHIV+ children aged 8-18 years and 37 controls who were matched for age, sex, ethnicity, and socioeconomic status. We explored associations between Lp(a) and disease- and treatment-related factors (inflammation, monocyte activation, and vascular), biomarkers, and neuroimaging outcomes using linear regression models. Results PHIV+ children had significantly higher levels of Lp(a) compared with controls (median, 43.6 [21.6-82.4] vs 21.8 [16.8-46.6] mg/dL; P = .033). Other lipid levels were comparable between groups. Additional assessment of apolipoprotein B, apolipoprotein CIII, apolipoprotein E, and APOE genotype revealed no significant differences. Higher Lp(a) levels were associated with higher plasma apoB levels and with lower monocyte chemoattractant protein-1 and TG levels in PHIV+ children. Lp(a) was not associated with HIV- or cART-related variables or with neuroimaging outcomes. Conclusions cART-treated PHIV+ children appear to have higher levels of Lp(a) compared with ethnicity-matched controls, which may implicate higher CVD risk in this population. Future research should focus on the association between Lp(a) and (sub)clinical CVD measurements in cART-treated PHIV+ patients. Dutch Trial Register number NRT4074.
Collapse
Affiliation(s)
- Malon Van den Hof
- Pediatric Infectious Diseases, Emma Children's Hospital, Amsterdam, the Netherlands
| | | | - Charlotte Blokhuis
- Pediatric Infectious Diseases, Emma Children's Hospital, Amsterdam, the Netherlands
| | | | - Hans P G Jansen
- Department of Experimental Vascular Medicine and Vascular Medicine, Amsterdam, the Netherlands
| | - Neeltje A Kootstra
- Department of Experimental Immunology, Amsterdam University Medical Centers, location Academic Medical Center, Amsterdam, the Netherlands
| | - Geesje M Dallinga-Thie
- Department of Experimental Vascular Medicine and Vascular Medicine, Amsterdam, the Netherlands
| | | | - Sotirios Tsimikas
- Sulpizio Cardiovascular Center, Division of Cardiovascular Medicine, University California San Diego, La Jolla, California
| | - Dasja Pajkrt
- Pediatric Infectious Diseases, Emma Children's Hospital, Amsterdam, the Netherlands
| | | |
Collapse
|
40
|
Tsimikas S, Fazio S, Ferdinand KC, Ginsberg HN, Koschinsky ML, Marcovina SM, Moriarty PM, Rader DJ, Remaley AT, Reyes-Soffer G, Santos RD, Thanassoulis G, Witztum JL, Danthi S, Olive M, Liu L. NHLBI Working Group Recommendations to Reduce Lipoprotein(a)-Mediated Risk of Cardiovascular Disease and Aortic Stenosis. J Am Coll Cardiol 2019; 71:177-192. [PMID: 29325642 DOI: 10.1016/j.jacc.2017.11.014] [Citation(s) in RCA: 329] [Impact Index Per Article: 65.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/02/2017] [Accepted: 11/06/2017] [Indexed: 12/16/2022]
Abstract
Pathophysiological, epidemiological, and genetic studies provide strong evidence that lipoprotein(a) [Lp(a)] is a causal mediator of cardiovascular disease (CVD) and calcific aortic valve disease (CAVD). Specific therapies to address Lp(a)-mediated CVD and CAVD are in clinical development. Due to knowledge gaps, the National Heart, Lung, and Blood Institute organized a working group that identified challenges in fully understanding the role of Lp(a) in CVD/CAVD. These included the lack of research funding, inadequate experimental models, lack of globally standardized Lp(a) assays, and inadequate understanding of the mechanisms underlying current drug therapies on Lp(a) levels. Specific recommendations were provided to facilitate basic, mechanistic, preclinical, and clinical research on Lp(a); foster collaborative research and resource sharing; leverage expertise of different groups and centers with complementary skills; and use existing National Heart, Lung, and Blood Institute resources. Concerted efforts to understand Lp(a) pathophysiology, together with diagnostic and therapeutic advances, are required to reduce Lp(a)-mediated risk of CVD and CAVD.
Collapse
Affiliation(s)
- Sotirios Tsimikas
- Vascular Medicine Program, Sulpizio Cardiovascular Center, Division of Cardiology, Department of Medicine, University of California San Diego, La Jolla, California.
| | - Sergio Fazio
- Oregon Health & Science University, Portland, Oregon
| | | | - Henry N Ginsberg
- College of Physicians and Surgeons, Columbia University, New York, New York
| | - Marlys L Koschinsky
- Robarts Research Institute and Department of Physiology & Pharmacology Schulich School of Medicine & Dentistry, University of Western Ontario, London, Ontario, Canada
| | | | | | - Daniel J Rader
- Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Alan T Remaley
- National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland
| | | | - Raul D Santos
- Heart Institute (InCor) University of Sao Paulo Medical School Hospital and Hospital Israelita Albert Einstein, Sao Paulo, Brazil
| | | | - Joseph L Witztum
- Division of Endocrinology, Department of Medicine, University of California San Diego, La Jolla, California
| | - Simhan Danthi
- National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland
| | - Michelle Olive
- National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland
| | - Lijuan Liu
- National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland
| |
Collapse
|
41
|
Satoh K, Shimokawa H. Recent Advances in the Development of Cardiovascular Biomarkers. Arterioscler Thromb Vasc Biol 2019; 38:e61-e70. [PMID: 29695533 DOI: 10.1161/atvbaha.118.310226] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Affiliation(s)
- Kimio Satoh
- From the Department of Cardiovascular Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Hiroaki Shimokawa
- From the Department of Cardiovascular Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan.
| |
Collapse
|
42
|
Blanchard V, Croyal M, Khantalin I, Ramin-Mangata S, Chemello K, Nativel B, Blom DJ, Marais AD, Lambert G. Reduced Lipoprotein(a) Associated With the Apolipoprotein E2 Genotype Confers Cardiovascular Protection in Familial Hypercholesterolemia. JACC Basic Transl Sci 2019; 4:425-427. [PMID: 31312765 PMCID: PMC6610154 DOI: 10.1016/j.jacbts.2019.03.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
43
|
Edfors F, Forsström B, Vunk H, Kotol D, Fredolini C, Maddalo G, Svensson AS, Boström T, Tegel H, Nilsson P, Schwenk JM, Uhlen M. Screening a Resource of Recombinant Protein Fragments for Targeted Proteomics. J Proteome Res 2019; 18:2706-2718. [PMID: 31094526 DOI: 10.1021/acs.jproteome.8b00924] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
The availability of proteomics resources hosting protein and peptide standards, as well as the data describing their analytical performances, will continue to enhance our current capabilities to develop targeted proteomics methods for quantitative biology. This study describes the analysis of a resource of 26,840 individually purified recombinant protein fragments corresponding to more than 16,000 human protein-coding genes. The resource was screened to identify proteotypic peptides suitable for targeted proteomics efforts, and we report LC-MS/MS assay coordinates for more than 25,000 proteotypic peptides, corresponding to more than 10,000 unique proteins. Additionally, peptide formation and digestion kinetics were, for a subset of the standards, monitored using a time-course protocol involving parallel digestion of isotope-labeled recombinant protein standards and endogenous human plasma proteins. We show that the strategy by adding isotope-labeled recombinant proteins before trypsin digestion enables short digestion protocols (≤60 min) with robust quantitative precision. In a proof-of-concept study, we quantified 23 proteins in human plasma using assay parameters defined in our study and used the standards to describe distinct clusters of individuals linked to different levels of LPA, APOE, SERPINA5, and TFRC. In summary, we describe the use and utility of a resource of recombinant proteins to identify proteotypic peptides useful for targeted proteomics assay development.
Collapse
Affiliation(s)
- Fredrik Edfors
- Science for Life Laboratory, Division of Systems Biology, Department of Protein Science , KTH-Royal Institute of Technology , SE - 171 21 Stockholm , Sweden
| | - Björn Forsström
- Science for Life Laboratory, Division of Systems Biology, Department of Protein Science , KTH-Royal Institute of Technology , SE - 171 21 Stockholm , Sweden
| | - Helian Vunk
- Science for Life Laboratory, Division of Systems Biology, Department of Protein Science , KTH-Royal Institute of Technology , SE - 171 21 Stockholm , Sweden
| | - David Kotol
- Science for Life Laboratory, Division of Systems Biology, Department of Protein Science , KTH-Royal Institute of Technology , SE - 171 21 Stockholm , Sweden
| | - Claudia Fredolini
- Science for Life Laboratory, Division of Affinity Proteomics, Department of Protein Science , KTH-Royal Institute of Technology , SE - 171 21 Stockholm , Sweden
| | - Gianluca Maddalo
- Science for Life Laboratory, Division of Systems Biology, Department of Protein Science , KTH-Royal Institute of Technology , SE - 171 21 Stockholm , Sweden
| | - Anne-Sophie Svensson
- Albanova University Center , KTH-Royal Institute of Technology , SE - 171 21 Stockholm , Sweden
| | - Tove Boström
- Albanova University Center , KTH-Royal Institute of Technology , SE - 171 21 Stockholm , Sweden.,Atlas Antibodies AB , SE - 114 21 Stockholm , Sweden
| | - Hanna Tegel
- Albanova University Center , KTH-Royal Institute of Technology , SE - 171 21 Stockholm , Sweden
| | - Peter Nilsson
- Science for Life Laboratory, Division of Affinity Proteomics, Department of Protein Science , KTH-Royal Institute of Technology , SE - 171 21 Stockholm , Sweden
| | - Jochen M Schwenk
- Science for Life Laboratory, Division of Affinity Proteomics, Department of Protein Science , KTH-Royal Institute of Technology , SE - 171 21 Stockholm , Sweden
| | - Mathias Uhlen
- Science for Life Laboratory, Division of Systems Biology, Department of Protein Science , KTH-Royal Institute of Technology , SE - 171 21 Stockholm , Sweden.,Albanova University Center , KTH-Royal Institute of Technology , SE - 171 21 Stockholm , Sweden.,Department of Neuroscience - Karolinska Institute , SE - 171 65 Solna , Sweden.,Novo Nordisk Foundation Center for Biosustainability , Technical University of Denmark , DK - 2970 Hørsholm , Denmark
| |
Collapse
|
44
|
Tsimikas S, Gordts PLSM, Nora C, Yeang C, Witztum JL. Statin therapy increases lipoprotein(a) levels. Eur Heart J 2019; 41:2275-2284. [DOI: 10.1093/eurheartj/ehz310] [Citation(s) in RCA: 165] [Impact Index Per Article: 33.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2018] [Revised: 09/18/2018] [Accepted: 04/27/2019] [Indexed: 01/03/2023] Open
Abstract
Abstract
Aims
Lipoprotein(a) [Lp(a)] is elevated in 20–30% of people. This study aimed to assess the effect of statins on Lp(a) levels.
Methods and results
This subject-level meta-analysis includes 5256 patients (1371 on placebo and 3885 on statin) from six randomized trials, three statin-vs.-placebo trials, and three statin-vs.-statin trials, with pre- and on-treatment (4–104 weeks) Lp(a) levels. Statins included atorvastatin 10 mg/day and 80 mg/day, pravastatin 40 mg/day, rosuvastatin 40 mg/day, and pitavastatin 2 mg/day. Lipoprotein(a) levels were measured with the same validated assay. The primary analysis of Lp(a) is based on the log-transformed data. In the statin-vs.-placebo pooled analysis, the ratio of geometric means [95% confidence interval (CI)] for statin to placebo is 1.11 (1.07–1.14) (P < 0.0001), with ratio >1 indicating a higher increase in Lp(a) from baseline in statin vs. placebo. The mean percent change from baseline ranged from 8.5% to 19.6% in the statin groups and −0.4% to −2.3% in the placebo groups. In the statin-vs.-statin pooled analysis, the ratio of geometric means (95% CI) for atorvastatin to pravastatin is 1.09 (1.05–1.14) (P < 0.0001). The mean percent change from baseline ranged from 11.6% to 20.4% in the pravastatin group and 18.7% to 24.2% in the atorvastatin group. Incubation of HepG2 hepatocytes with atorvastatin showed an increase in expression of LPA mRNA and apolipoprotein(a) protein.
Conclusion
This meta-analysis reveals that statins significantly increase plasma Lp(a) levels. Elevations of Lp(a) post-statin therapy should be studied for effects on residual cardiovascular risk.
Collapse
Affiliation(s)
- Sotirios Tsimikas
- Vascular Medicine Program, Sulpizio Cardiovascular Center, Division of Cardiovascular Diseases, University of California San Diego, 9500 Gilman Drive, La Jolla, CA 92093-0682, USA
| | - Philip L S M Gordts
- Department of Cellular and Molecular Medicine, Glycobiology Research and Training Center, University of California San Diego, 9500 Gilman Drive, La Jolla, CA 92093-0682, USA
- Department of Medicine, Division of Endocrinology and Metabolism, University of California San Diego, 9500 Gilman Drive, La Jolla, CA 92093-0682, USA
| | - Chelsea Nora
- Department of Cellular and Molecular Medicine, Glycobiology Research and Training Center, University of California San Diego, 9500 Gilman Drive, La Jolla, CA 92093-0682, USA
- Department of Medicine, Division of Endocrinology and Metabolism, University of California San Diego, 9500 Gilman Drive, La Jolla, CA 92093-0682, USA
| | - Calvin Yeang
- Vascular Medicine Program, Sulpizio Cardiovascular Center, Division of Cardiovascular Diseases, University of California San Diego, 9500 Gilman Drive, La Jolla, CA 92093-0682, USA
| | - Joseph L Witztum
- Department of Medicine, Division of Endocrinology and Metabolism, University of California San Diego, 9500 Gilman Drive, La Jolla, CA 92093-0682, USA
| |
Collapse
|
45
|
Parish S, Hopewell JC, Hill MR, Marcovina S, Valdes-Marquez E, Haynes R, Offer A, Pedersen TR, Baigent C, Collins R, Landray M, Armitage J. Impact of Apolipoprotein(a) Isoform Size on Lipoprotein(a) Lowering in the HPS2-THRIVE Study. CIRCULATION-GENOMIC AND PRECISION MEDICINE 2019; 11:e001696. [PMID: 29449329 PMCID: PMC5841847 DOI: 10.1161/circgen.117.001696] [Citation(s) in RCA: 63] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/28/2017] [Accepted: 12/01/2017] [Indexed: 12/28/2022]
Abstract
Supplemental Digital Content is available in the text. Background: Genetic studies have shown lipoprotein(a) (Lp[a]) to be an important causal risk factor for coronary disease. Apolipoprotein(a) isoform size is the chief determinant of Lp(a) levels, but its impact on the benefits of therapies that lower Lp(a) remains unclear. Methods: HPS2-THRIVE (Heart Protection Study 2–Treatment of HDL to Reduce the Incidence of Vascular Events) is a randomized trial of niacin–laropiprant versus placebo on a background of simvastatin therapy. Plasma Lp(a) levels at baseline and 1 year post-randomization were measured in 3978 participants from the United Kingdom and China. Apolipoprotein(a) isoform size, estimated by the number of kringle IV domains, was measured by agarose gel electrophoresis and the predominantly expressed isoform identified. Results: Allocation to niacin–laropiprant reduced mean Lp(a) by 12 (SE, 1) nmol/L overall and 34 (6) nmol/L in the top quintile by baseline Lp(a) level (Lp[a] ≥128 nmol/L). The mean proportional reduction in Lp(a) with niacin–laropiprant was 31% but varied strongly with predominant apolipoprotein(a) isoform size (PTrend=4×10−29) and was only 18% in the quintile with the highest baseline Lp(a) level and low isoform size. Estimates from genetic studies suggest that these Lp(a) reductions during the short term of the trial might yield proportional reductions in coronary risk of ≈2% overall and 6% in the top quintile by Lp(a) levels. Conclusions: Proportional reductions in Lp(a) were dependent on apolipoprotein(a) isoform size. Taking this into account, the likely benefits of niacin–laropiprant on coronary risk through Lp(a) lowering are small. Novel therapies that reduce high Lp(a) levels by at least 80 nmol/L (≈40%) may be needed to produce worthwhile benefits in people at the highest risk because of Lp(a). Clinical Trial Registration: URL: https://clinicaltrials.gov. Unique identifier: NCT00461630.
Collapse
Affiliation(s)
- Sarah Parish
- From the Medical Research Council Population Health Research Unit (S.P., M.R.H., R.H., C.B., J.A.); and the Clinical Trial Service Unit and Epidemiological Studies Unit (S.P., J.C.H., M.R.H., E.V.-M., R.H., A.O., C.B., R.C., M.L., J.A.), Nuffield Department of Population Health, University of Oxford, United Kingdom; Northwest Lipid Metabolism and Diabetes Research Laboratories, University of Washington, Seattle (S.M.); and Center for Preventive Medicine, University of Oslo, Norway (T.R.P.). A complete list of collaborators in HPS2-THRIVE (Heart Protection Study 2-Treatment of HDL to Reduce the Incidence of Vascular Events) is given in reference 13.
| | - Jemma C Hopewell
- From the Medical Research Council Population Health Research Unit (S.P., M.R.H., R.H., C.B., J.A.); and the Clinical Trial Service Unit and Epidemiological Studies Unit (S.P., J.C.H., M.R.H., E.V.-M., R.H., A.O., C.B., R.C., M.L., J.A.), Nuffield Department of Population Health, University of Oxford, United Kingdom; Northwest Lipid Metabolism and Diabetes Research Laboratories, University of Washington, Seattle (S.M.); and Center for Preventive Medicine, University of Oslo, Norway (T.R.P.). A complete list of collaborators in HPS2-THRIVE (Heart Protection Study 2-Treatment of HDL to Reduce the Incidence of Vascular Events) is given in reference 13
| | - Michael R Hill
- From the Medical Research Council Population Health Research Unit (S.P., M.R.H., R.H., C.B., J.A.); and the Clinical Trial Service Unit and Epidemiological Studies Unit (S.P., J.C.H., M.R.H., E.V.-M., R.H., A.O., C.B., R.C., M.L., J.A.), Nuffield Department of Population Health, University of Oxford, United Kingdom; Northwest Lipid Metabolism and Diabetes Research Laboratories, University of Washington, Seattle (S.M.); and Center for Preventive Medicine, University of Oslo, Norway (T.R.P.). A complete list of collaborators in HPS2-THRIVE (Heart Protection Study 2-Treatment of HDL to Reduce the Incidence of Vascular Events) is given in reference 13
| | - Santica Marcovina
- From the Medical Research Council Population Health Research Unit (S.P., M.R.H., R.H., C.B., J.A.); and the Clinical Trial Service Unit and Epidemiological Studies Unit (S.P., J.C.H., M.R.H., E.V.-M., R.H., A.O., C.B., R.C., M.L., J.A.), Nuffield Department of Population Health, University of Oxford, United Kingdom; Northwest Lipid Metabolism and Diabetes Research Laboratories, University of Washington, Seattle (S.M.); and Center for Preventive Medicine, University of Oslo, Norway (T.R.P.). A complete list of collaborators in HPS2-THRIVE (Heart Protection Study 2-Treatment of HDL to Reduce the Incidence of Vascular Events) is given in reference 13
| | - Elsa Valdes-Marquez
- From the Medical Research Council Population Health Research Unit (S.P., M.R.H., R.H., C.B., J.A.); and the Clinical Trial Service Unit and Epidemiological Studies Unit (S.P., J.C.H., M.R.H., E.V.-M., R.H., A.O., C.B., R.C., M.L., J.A.), Nuffield Department of Population Health, University of Oxford, United Kingdom; Northwest Lipid Metabolism and Diabetes Research Laboratories, University of Washington, Seattle (S.M.); and Center for Preventive Medicine, University of Oslo, Norway (T.R.P.). A complete list of collaborators in HPS2-THRIVE (Heart Protection Study 2-Treatment of HDL to Reduce the Incidence of Vascular Events) is given in reference 13
| | - Richard Haynes
- From the Medical Research Council Population Health Research Unit (S.P., M.R.H., R.H., C.B., J.A.); and the Clinical Trial Service Unit and Epidemiological Studies Unit (S.P., J.C.H., M.R.H., E.V.-M., R.H., A.O., C.B., R.C., M.L., J.A.), Nuffield Department of Population Health, University of Oxford, United Kingdom; Northwest Lipid Metabolism and Diabetes Research Laboratories, University of Washington, Seattle (S.M.); and Center for Preventive Medicine, University of Oslo, Norway (T.R.P.). A complete list of collaborators in HPS2-THRIVE (Heart Protection Study 2-Treatment of HDL to Reduce the Incidence of Vascular Events) is given in reference 13
| | - Alison Offer
- From the Medical Research Council Population Health Research Unit (S.P., M.R.H., R.H., C.B., J.A.); and the Clinical Trial Service Unit and Epidemiological Studies Unit (S.P., J.C.H., M.R.H., E.V.-M., R.H., A.O., C.B., R.C., M.L., J.A.), Nuffield Department of Population Health, University of Oxford, United Kingdom; Northwest Lipid Metabolism and Diabetes Research Laboratories, University of Washington, Seattle (S.M.); and Center for Preventive Medicine, University of Oslo, Norway (T.R.P.). A complete list of collaborators in HPS2-THRIVE (Heart Protection Study 2-Treatment of HDL to Reduce the Incidence of Vascular Events) is given in reference 13
| | - Terje R Pedersen
- From the Medical Research Council Population Health Research Unit (S.P., M.R.H., R.H., C.B., J.A.); and the Clinical Trial Service Unit and Epidemiological Studies Unit (S.P., J.C.H., M.R.H., E.V.-M., R.H., A.O., C.B., R.C., M.L., J.A.), Nuffield Department of Population Health, University of Oxford, United Kingdom; Northwest Lipid Metabolism and Diabetes Research Laboratories, University of Washington, Seattle (S.M.); and Center for Preventive Medicine, University of Oslo, Norway (T.R.P.). A complete list of collaborators in HPS2-THRIVE (Heart Protection Study 2-Treatment of HDL to Reduce the Incidence of Vascular Events) is given in reference 13
| | - Colin Baigent
- From the Medical Research Council Population Health Research Unit (S.P., M.R.H., R.H., C.B., J.A.); and the Clinical Trial Service Unit and Epidemiological Studies Unit (S.P., J.C.H., M.R.H., E.V.-M., R.H., A.O., C.B., R.C., M.L., J.A.), Nuffield Department of Population Health, University of Oxford, United Kingdom; Northwest Lipid Metabolism and Diabetes Research Laboratories, University of Washington, Seattle (S.M.); and Center for Preventive Medicine, University of Oslo, Norway (T.R.P.). A complete list of collaborators in HPS2-THRIVE (Heart Protection Study 2-Treatment of HDL to Reduce the Incidence of Vascular Events) is given in reference 13
| | - Rory Collins
- From the Medical Research Council Population Health Research Unit (S.P., M.R.H., R.H., C.B., J.A.); and the Clinical Trial Service Unit and Epidemiological Studies Unit (S.P., J.C.H., M.R.H., E.V.-M., R.H., A.O., C.B., R.C., M.L., J.A.), Nuffield Department of Population Health, University of Oxford, United Kingdom; Northwest Lipid Metabolism and Diabetes Research Laboratories, University of Washington, Seattle (S.M.); and Center for Preventive Medicine, University of Oslo, Norway (T.R.P.). A complete list of collaborators in HPS2-THRIVE (Heart Protection Study 2-Treatment of HDL to Reduce the Incidence of Vascular Events) is given in reference 13
| | - Martin Landray
- From the Medical Research Council Population Health Research Unit (S.P., M.R.H., R.H., C.B., J.A.); and the Clinical Trial Service Unit and Epidemiological Studies Unit (S.P., J.C.H., M.R.H., E.V.-M., R.H., A.O., C.B., R.C., M.L., J.A.), Nuffield Department of Population Health, University of Oxford, United Kingdom; Northwest Lipid Metabolism and Diabetes Research Laboratories, University of Washington, Seattle (S.M.); and Center for Preventive Medicine, University of Oslo, Norway (T.R.P.). A complete list of collaborators in HPS2-THRIVE (Heart Protection Study 2-Treatment of HDL to Reduce the Incidence of Vascular Events) is given in reference 13
| | - Jane Armitage
- From the Medical Research Council Population Health Research Unit (S.P., M.R.H., R.H., C.B., J.A.); and the Clinical Trial Service Unit and Epidemiological Studies Unit (S.P., J.C.H., M.R.H., E.V.-M., R.H., A.O., C.B., R.C., M.L., J.A.), Nuffield Department of Population Health, University of Oxford, United Kingdom; Northwest Lipid Metabolism and Diabetes Research Laboratories, University of Washington, Seattle (S.M.); and Center for Preventive Medicine, University of Oslo, Norway (T.R.P.). A complete list of collaborators in HPS2-THRIVE (Heart Protection Study 2-Treatment of HDL to Reduce the Incidence of Vascular Events) is given in reference 13
| | | |
Collapse
|
46
|
Chan DC, Watts GF, Coll B, Wasserman SM, Marcovina SM, Barrett PHR. Lipoprotein(a) Particle Production as a Determinant of Plasma Lipoprotein(a) Concentration Across Varying Apolipoprotein(a) Isoform Sizes and Background Cholesterol-Lowering Therapy. J Am Heart Assoc 2019; 8:e011781. [PMID: 30897995 PMCID: PMC6509712 DOI: 10.1161/jaha.118.011781] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/25/2018] [Accepted: 02/12/2019] [Indexed: 12/24/2022]
Abstract
Background Elevated lipoprotein(a) (Lp(a)), a low-density lipoprotein-like particle bound to the polymorphic apolipoprotein(a) (apo(a)), may be causal for cardiovascular disease. However, the metabolism of Lp(a) in humans is poorly understood. Methods and Results We investigated the kinetics of Lp(a)-apo(a) and low-density lipoprotein-apoB-100 in 63 normolipidemic men. The fractional catabolic rate ( FCR ) and production rate PR ) were studied. Plasma apo(a) concentration was significantly and inversely associated with apo(a) isoform size ( r=-0.536, P<0.001) and apo(a) FCR ( r=-0.363, P<0.01), and positively with apo(a) PR ( r=0.877, P<0.001). There were no significant associations between the FCR s of apo(a) and low-density lipoprotein-apoB-100. Subjects with smaller apo(a) isoform sizes (≤22 kringle IV repeats) had significantly higher apo(a) PR ( P<0.05) and lower apo(a) FCR ( P<0.01) than those with larger sizes. Plasma apo(a) concentration was significantly associated with apo(a) PR ( r=0.930, P<0.001), but not with FCR ( r=-0.012, P>0.05) in subjects with smaller apo(a) isoform size. In contrast, both apo(a) PR and FCR were significantly associated with plasma apo(a) concentrations ( r=0.744 and -0.389, respectively, P<0.05) in subjects with larger isoforms. In multiple regression analysis, apo(a) PR and apo(a) isoform size were significant predictors of plasma apo(a) concentration independent of low-density lipoprotein-apoB-100 FCR and background therapy with atorvastatin and evolocumab. Conclusions In normolipidemic men, the plasma Lp(a) concentration is predominantly determined by the rate of production of Lp(a) particles, irrespective of apo(a) isoform size and background therapy with a statin and a proprotein convertase subtilisin-kexin type 9 inhibitor. Our findings underscore the importance of therapeutic targeting of the hepatic synthesis and secretion of Lp(a) particles. Lp(a) particle catabolism may only play a modest role in determining Lp(a) concentration in subjects with larger apo(a) isoform size. Clinical Trial Registration URL : http://www.clinicaltrials.gov . Unique identifier: NCT 02189837.
Collapse
Affiliation(s)
- Dick C. Chan
- School of MedicineUniversity of Western AustraliaPerthAustralia
- School of Biomedical ScienceUniversity of Western AustraliaPerthAustralia
| | - Gerald F. Watts
- School of MedicineUniversity of Western AustraliaPerthAustralia
- The Lipid Disorders ClinicDepartment of CardiologyRoyal Perth HospitalPerthAustralia
| | | | | | - Santica M. Marcovina
- Northwest Lipid Metabolism and Diabetes Research LaboratoriesDivision of Metabolism, Endocrinology, and NutritionDepartment of MedicineUniversity of WashingtonSeattleWA
| | - P. Hugh R. Barrett
- School of Biomedical ScienceUniversity of Western AustraliaPerthAustralia
- Faculty of Medicine and HealthUniversity of New EnglandArmidaleNew South WalesAustralia
| |
Collapse
|
47
|
Reiner Ž. Can Lp(a) Lowering Against Background Statin Therapy Really Reduce Cardiovascular Risk? Curr Atheroscler Rep 2019; 21:14. [PMID: 30847681 DOI: 10.1007/s11883-019-0773-y] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
PURPOSE OF REVIEW The association between elevated plasma levels of lipoprotein (a) [Lp(a)] and atherosclerotic cardiovascular disease (ASCVD) has been discussed for many years. Recent genetic findings have confirmed that elevated Lp(a) similar to elevated LDL-cholesterol (LDL-C) might be causally related to premature ASCVD. Lp(a) is relatively refractory to lifestyle interventions. The results of studies with statins and their possible effect on Lp(a) are conflicting. Specific Lp(a) apheresis is used as a treatment against background statin therapy and can decrease Lp(a). The purpose of this review is to discuss whether new drugs which decrease Lp(a) can prevent ASCVD and decrease ASCVD mortality when applied in addition to statins. RECENT FINDINGS Some new LDL-C-lowering drugs such as mipomersen and lomitapide decrease elevated Lp(a) in addition to statins but they have some unpleasant adverse effects. Recently, an antisense oligonucleotide against apo(a), AKCEA-APO(a)Rx, has been shown to selectively decrease Lp(a). The most recent advance in LDL-C lowering are PCSK9 inhibitors. Alirocumab and evolocumab do not only significantly reduce LDL-C on top of maximally tolerated statin therapy and prevent ASCVD events, but also further decrease Lp(a). There is no data to indicate whether mipomersen, lomitapide, or IONIS-APO(a)-LRx decrease ASCVD events and mortality. Conclusive evidence is still lacking as to whether the treatment with PCSK9 inhibitors against background statin therapy actually additionally reduces ASCVD risk due to the lowering of Lp(a) or simply due to lowering LDL-C to levels much lower than high-intensity statin treatment as monotherapy. Ongoing trials will probably provide an answer to these questions.
Collapse
Affiliation(s)
- Željko Reiner
- Department of Internal Medicine, University Hospital Centre Zagreb, School of Medicine, University of Zagreb, Kispaticeva 12, 10000, Zagreb, Croatia.
| |
Collapse
|
48
|
Bea AM, Lamiquiz-Moneo I, Marco-Benedí V, Mateo-Gallego R, Pérez-Calahorra S, Jarauta E, Martín C, Cenarro A, Civeira F. Lipid-lowering response in subjects with the p.(Leu167del) mutation in the APOE gene. Atherosclerosis 2019; 282:143-147. [DOI: 10.1016/j.atherosclerosis.2019.01.024] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2018] [Revised: 01/09/2019] [Accepted: 01/16/2019] [Indexed: 01/09/2023]
|
49
|
Abstract
The organs require oxygen and other types of nutrients (amino acids, sugars, and lipids) to function, the heart consuming large amounts of fatty acids for oxidation and adenosine triphosphate (ATP) generation.
Collapse
|
50
|
Matsuura Y, Kanter JE, Bornfeldt KE. Highlighting Residual Atherosclerotic Cardiovascular Disease Risk. Arterioscler Thromb Vasc Biol 2019; 39:e1-e9. [PMID: 30586334 PMCID: PMC6310032 DOI: 10.1161/atvbaha.118.311999] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Affiliation(s)
- Yunosuke Matsuura
- From the Department of Medicine, University of Washington Medicine Diabetes Institute, University of Washington School of Medicine, Seattle (Y.M., J.E.K., K.E.B.)
| | - Jenny E Kanter
- From the Department of Medicine, University of Washington Medicine Diabetes Institute, University of Washington School of Medicine, Seattle (Y.M., J.E.K., K.E.B.)
| | - Karin E Bornfeldt
- From the Department of Medicine, University of Washington Medicine Diabetes Institute, University of Washington School of Medicine, Seattle (Y.M., J.E.K., K.E.B.)
- Department of Pathology, University of Washington Medicine Diabetes Institute, University of Washington School of Medicine, Seattle (K.E.B.)
| |
Collapse
|