51
|
Koschinsky ML, Boffa MB. Lipoprotein(a): an important cardiovascular risk factor and a clinical conundrum. Endocrinol Metab Clin North Am 2014; 43:949-62. [PMID: 25432390 DOI: 10.1016/j.ecl.2014.08.002] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Elevated plasma concentrations of lipoprotein(a) (Lp[a]) are an emerging risk factor for the development of coronary heart disease (CHD). Recent genetic and epidemiologic data have provided strong evidence for a causal role of Lp(a) in CHD. Despite these developments, which have attracted increasing interest from clinicians and basic scientists, many unanswered questions persist. The true pathogenic mechanism of Lp(a) remains a mystery. Significant uncertainty exists concerning the appropriate use of Lp(a) in the clinical setting. No therapeutic intervention remains that can specifically lower plasma Lp(a) concentrations, although the list of compounds that lower Lp(a) and LDL continues to expand.
Collapse
Affiliation(s)
- Marlys L Koschinsky
- Department of Chemistry and Biochemistry, University of Windsor, 401 Sunset Avenue, Windsor, Ontario N9B 3P4, Canada.
| | - Michael B Boffa
- Department of Chemistry and Biochemistry, University of Windsor, 401 Sunset Avenue, Windsor, Ontario N9B 3P4, Canada
| |
Collapse
|
52
|
Affiliation(s)
- Stephen J Nicholls
- South Australian Health and Medical Research Institute, University of South Australia, Adelaide, Australia; Discipline of Medicine, University of Adelaide, Adelaide, Australia.
| | - Alex Brown
- South Australian Health and Medical Research Institute, University of South Australia, Adelaide, Australia; School of Population Health, University of South Australia, Adelaide, Australia
| |
Collapse
|
53
|
Effect of alirocumab, a monoclonal proprotein convertase subtilisin/kexin 9 antibody, on lipoprotein(a) concentrations (a pooled analysis of 150 mg every two weeks dosing from phase 2 trials). Am J Cardiol 2014; 114:711-5. [PMID: 25060413 DOI: 10.1016/j.amjcard.2014.05.060] [Citation(s) in RCA: 158] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/24/2014] [Revised: 05/28/2014] [Accepted: 05/28/2014] [Indexed: 11/23/2022]
Abstract
Lipoprotein(a) [Lp(a)] is an independent risk factor for cardiovascular disease, with limited treatment options. This analysis evaluated the effect of a monoclonal antibody to proprotein convertase subtilisin/kexin 9, alirocumab 150 mg every 2 weeks (Q2W), on Lp(a) levels in pooled data from 3 double-blind, randomized, placebo-controlled, phase 2 studies of 8 or 12 weeks' duration conducted in patients with hypercholesterolemia on background lipid-lowering therapy (NCT01266876, NCT01288469, and NCT01288443). Data were available for 102 of 108 patients who received alirocumab 150 mg Q2W and 74 of 77 patients who received placebo. Alirocumab resulted in a significant reduction in Lp(a) from baseline compared with placebo (-30.3% vs -0.3%, p <0.0001). Median percentage Lp(a) reductions in the alirocumab group were of a similar magnitude across a range of baseline Lp(a) levels, resulting in greater absolute reductions in Lp(a) in patients with higher baseline levels. Regression analysis indicated that <5% of the variance in the reduction of Lp(a) was explained by the effect of alirocumab on low-density lipoprotein cholesterol. In conclusion, pooled data from 3 phase 2 trials demonstrate substantive reduction in Lp(a) with alirocumab 150 mg Q2W, including patients with baseline Lp(a) >50 mg/dl. Reductions in Lp(a) only weakly correlated with the magnitude of low-density lipoprotein cholesterol lowering.
Collapse
|
54
|
Ezhov MV, Safarova MS, Afanasieva OI, Kukharchuk VV, Pokrovsky SN. Lipoprotein(a) level and apolipoprotein(a) phenotype as predictors of long-term cardiovascular outcomes after coronary artery bypass grafting. Atherosclerosis 2014; 235:477-82. [PMID: 24952151 DOI: 10.1016/j.atherosclerosis.2014.05.944] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/30/2013] [Revised: 05/19/2014] [Accepted: 05/23/2014] [Indexed: 11/18/2022]
Abstract
OBJECTIVE To evaluate the relationships of lipoprotein(a) (Lp(a)) concentration and apolipoprotein(a) (apo(a)) phenotype to major adverse cardiovascular events after coronary artery bypass grafting (CABG) in long-term follow-up. METHODS This single-center study included 356 patients with stable coronary heart disease (CHD) who underwent successful CABG. At baseline, we assessed the patient's risk factor profile for atherosclerosis, Lp(a) concentration and apo(a) phenotype. The primary endpoint was the composite of cardiovascular death and non-fatal myocardial infarction (MI). The secondary endpoint also included hospitalization for recurrent or unstable angina and repeat revascularization. RESULTS Over a mean of 8.5 ± 3.5 years (range 0.9-15.0 years), the primary and secondary endpoints were registered in 46 (13%) and 107 (30%) patients, respectively. Patients with Lp(a) ≥30 mg/dL were at significantly greater risk for the primary endpoint (hazard ratio (HR) 2.98, 95% confidence interval (CI) 1.76-5.03, p < 0.001) and secondary endpoint (HR 3.47, 95% CI 2.48-4.85, p < 0.001) than patients with Lp(a) values <30 mg/dL. The low molecular-weight apo(a) phenotype was also associated with higher risk of both primary and secondary endpoints (3.57 (1.87-6.82) and 3.05 (2.00-4.62), respectively; p < 0.001 for both), regardless of conventional risk factors and statins use. CONCLUSION In stable CHD patients Lp(a) concentration and low molecular-weight apo(a) phenotype are independently associated with three-fold increase in risk of major adverse cardiovascular events within 15 years after CABG. Lp(a) levels may provide an additional information for postoperative cardiovascular risk assessment.
Collapse
Affiliation(s)
- Marat V Ezhov
- Atherosclerosis Department, Institute of Clinical Cardiology named after A.L. Myasnikov, Federal State Institution "Russian Cardiology Research and Production Complex" of Ministry of Health of the Russian Federation, 15A, 3rd Cherepkovskaya street, Moscow 121552, Russia.
| | - Maya S Safarova
- Atherosclerosis Department, Institute of Clinical Cardiology named after A.L. Myasnikov, Federal State Institution "Russian Cardiology Research and Production Complex" of Ministry of Health of the Russian Federation, 15A, 3rd Cherepkovskaya street, Moscow 121552, Russia.
| | - Olga I Afanasieva
- Laboratory of Atherosclerosis, Institute of Experimental Cardiology, Federal State Institution "Russian Cardiology Research and Production Complex" of Ministry of Health of the Russian Federation, 15A, 3rd Cherepkovskaya street, Moscow 121552, Russia.
| | - Valery V Kukharchuk
- Atherosclerosis Department, Institute of Clinical Cardiology named after A.L. Myasnikov, Federal State Institution "Russian Cardiology Research and Production Complex" of Ministry of Health of the Russian Federation, 15A, 3rd Cherepkovskaya street, Moscow 121552, Russia.
| | - Sergei N Pokrovsky
- Laboratory of Atherosclerosis, Institute of Experimental Cardiology, Federal State Institution "Russian Cardiology Research and Production Complex" of Ministry of Health of the Russian Federation, 15A, 3rd Cherepkovskaya street, Moscow 121552, Russia.
| |
Collapse
|
55
|
Nozue T, Yamamoto S, Tohyama S, Fukui K, Umezawa S, Onishi Y, Kunishima T, Sato A, Nozato T, Miyake S, Takeyama Y, Morino Y, Yamauchi T, Muramatsu T, Hibi K, Terashima M, Michishita I. Lipoprotein(a) is associated with necrotic core progression of non-culprit coronary lesions in statin-treated patients with angina pectoris. Lipids Health Dis 2014; 13:59. [PMID: 24684829 PMCID: PMC4230650 DOI: 10.1186/1476-511x-13-59] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2014] [Accepted: 03/18/2014] [Indexed: 01/23/2023] Open
Abstract
Background Statin therapy results in regression and stabilization of coronary artery plaques, and reduces the incidence of coronary artery disease. However, statin therapy does not effectively halt the accumulation of necrotic core in all patients. The purpose of the present study was to identify the predictors associated with necrotic core progression during statin therapy. Methods Coronary atherosclerosis in non-culprit lesions was evaluated using virtual histology intravascular ultrasound at baseline and 8 months after statin therapy. One hundred nineteen patients were divided into 2 groups based on necrotic core progression or regression during an 8-month follow-up period. Results Patients with necrotic core progression had higher serum lipoprotein(a) [Lp(a)] levels than patients with regression at baseline (16 mg/dL vs. 12 mg/dL, p = 0.02) and at the 8-month follow-up (17 mg/dL vs. 10 mg/dL, p = 0.006). Patients with necrotic core progression had a higher fibro-fatty plaque volume (1.28 mm3/mm vs. 0.73 mm3/mm, p = 0.002), and less necrotic core (0.56 mm3/mm vs. 1.04 mm3/mm, p < 0.0001) and dense calcium (0.35 mm3/mm vs. 0.56 mm3/mm, p = 0.006) plaque volumes at baseline than patients with regression. Multivariate logistic regression analysis showed that Lp(a) was a significant independent predictor associated with necrotic core progression during statin therapy (odds ratio [OR]: 3.514; 95% confidence interval [CI]: 1.338-9.228; p = 0.01). Conclusions Serum Lp(a) is independently associated with necrotic core progression in statin-treated patients with angina pectoris.
Collapse
Affiliation(s)
- Tsuyoshi Nozue
- Division of Cardiology, Department of Internal Medicine, Yokohama Sakae Kyosai Hospital, 132 Katsura-cho, Sakae-ku, Yokohama 247-8581, Japan.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
56
|
The association between the LPA gene polymorphism and coronary artery disease in Chinese Han population. BIOMED RESEARCH INTERNATIONAL 2014; 2014:370670. [PMID: 24790998 PMCID: PMC3984839 DOI: 10.1155/2014/370670] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 01/23/2014] [Accepted: 02/26/2014] [Indexed: 01/13/2023]
Abstract
Lp(a) has been well known as an independent risk factor for coronary artery disease (CAD). The LPA gene, as it encodes apo(a) of the Lp(a) lipoprotein particle, was associated with increased risk of CAD. The purpose of this study was to analyze the relationship between the polymorphisms of LPA gene and CAD in Chinese Han population. Five SNPs (rs1367211, rs3127596, rs6415085, rs9347438, and rs9364559) in the LPA gene were genotyped using Sequenom MassARRAY time-of-flight mass spectrometer (TOF) in 560 CAD patients as case group and 531 non-CAD subjects as control group. The numbers of these two groups were from Chinese Han ancestry. The results showed that allele (P = 0.046) and genotype (P = 0.026) of rs9364559 in the LPA gene was associated with CAD. The frequency of rs9364559 minor allele (G) in case group was obviously higher than that in control group. Results of haplotype analysis showed that 4 haplotypes which contained rs9364559-G were associated with increased risk of CAD in this population. This study explored rs9364559 in the LPA gene may be associated with the pathogenesis of CAD; and the risk of CAD might be higher in the population carrying 4 haplotypes of different blocks in the LPA gene.
Collapse
|
57
|
Calmarza P, Bajador E, Lapresta C, García Castañón S, de Castro I, Civeira F. [Effect of biliary obstruction on lipoprotein(a) concentration]. CLINICA E INVESTIGACION EN ARTERIOSCLEROSIS 2014; 26:218-23. [PMID: 24629889 DOI: 10.1016/j.arteri.2014.01.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/23/2013] [Revised: 01/29/2014] [Accepted: 01/30/2014] [Indexed: 10/25/2022]
Abstract
OBJECTIVES This study was appointed to determine the correlation between the concentration of lipoprotein(a) [Lp(a)], apolipoproteins and lipids with biochemical parameters of liver function in a group of patients with reversible cholestasis. We have also determined the concentration of these parameters once solved the biliary obstruction process. MATERIAL AND METHODS Eighteen adults over 17 years with extrahepatic cholestasis were included in the study on a prospective basis, and we determined in them biochemical liver function parameters and lipoprotein metabolism parameters, particularly Lp(a) before and after unblocking. RESULTS The concentration of Lp(a) prior to desobstruction was inverse and statistically significantly correlated with the concentration of gamma glutamyl transpeptidase (correlation coefficient [r] = -0.757, P = .018). The concentration of Lp(a) (median = 2.66 mg/dL, interquartile range = 5,62) showed a statistically significant increase (median = 9.72 mg/dL, interquartile range = 28.76, P < .001), once the unblocking was performed. Concentrations of total cholesterol and triglycerides had a statistically significant decrease, and HDL cholesterol and apolipoprotein A-1 showed a statistically significant increase once the unblocking was carried out. CONCLUSIONS The concentration of Lp(a) is decreased during cholestasis, although there is a significant simultaneous hypercholesterolemia. Cholestasis has a causal role in lowering Lp(a), because the unblocking of bile duct recovers Lp(a) concentration. Our study supports the concept that bile acids exert a controlling effect on the synthesis of Lp(a) and open a mechanism for the treatment of hyper Lp(a).
Collapse
Affiliation(s)
- Pilar Calmarza
- Servicio de Bioquímica Clínica, Hospital Universitario Miguel Servet, Zaragoza, España.
| | - Eduardo Bajador
- Servicio de Digestivo, Hospital Universitario Miguel Servet, Zaragoza, España
| | - Carlos Lapresta
- Servicio de Medicina Preventiva, Hospital Universitario Miguel Servet, Zaragoza, España
| | | | - Isabel de Castro
- Unidad de Lípidos y Laboratorio de Investigación Molecular, Hospital Universitario Miguel Servet, Zaragoza, España
| | - Fernando Civeira
- Unidad de Lípidos y Laboratorio de Investigación Molecular, Hospital Universitario Miguel Servet, Zaragoza, España
| |
Collapse
|
58
|
Baseline LDL-C and Lp(a) elevations portend a high risk of coronary revascularization in patients after stent placement. DISEASE MARKERS 2013; 35:857-62. [PMID: 24367139 PMCID: PMC3866824 DOI: 10.1155/2013/472845] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 08/14/2013] [Revised: 10/29/2013] [Accepted: 11/03/2013] [Indexed: 01/07/2023]
Abstract
Background and Aim. Incidence of coronary restenosis after stent placement is high. Our study was going to investigate whether Lp(a) elevation was potential for predicting coronary restenosis and whether the effects of Lp(a) elevation on coronary restenosis were dependent on LDL-C level. Methods and Results. Totally 832 participants eligible for stent placement were enrolled and followed up for monitoring clinical end points. Baseline characteristics were collected. According to the cut point of Lp(a), participants were divided into low Lp(a) group (Lp(a) < 30 mg/dL) and high Lp(a) group (Lp(a) ≥ 30 mg/dL). Furthermore, based on baseline LDL-C level, participants were divided into LDL-C < 1.8 mmol/L and ≥1.8 mmol/L subgroups. Clinical end points including major adverse cardiovascular events (MACE), cardiovascular death, nonfatal myocardial infarction, ischemic stroke, and coronary revascularization (CR) were compared. Patients in high Lp(a) groups more frequently presented with acute coronary syndrome and three vessel stenoses. In subgroup of LDL-C < 1.8 mmol/L, no significant differences of cardiovascular outcomes were found between low and high Lp(a) groups. While in the subgroup of LDL-C ≥ 1.8 mmol/L, incidences of MACE and CR were significantly higher in high Lp(a) group, and odds ratio for CR was 2.05. Conclusion. With baseline LDL-C and Lp(a) elevations, incidence of CR is significantly increased after stent placement.
Collapse
|
59
|
Nestel PJ, Barnes EH, Tonkin AM, Simes J, Fournier M, White HD, Colquhoun DM, Blankenberg S, Sullivan DR. Plasma Lipoprotein(a) Concentration Predicts Future Coronary and Cardiovascular Events in Patients With Stable Coronary Heart Disease. Arterioscler Thromb Vasc Biol 2013; 33:2902-8. [DOI: 10.1161/atvbaha.113.302479] [Citation(s) in RCA: 92] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Affiliation(s)
- Paul J. Nestel
- From the Lipoprotein and Atherosclerosis Laboratory, Baker IDI Heart & Diabetes Institute, Melbourne, Australia (P.J.N.); NHMRC Clinical Trials Centre, University of Sydney, Sydney, Australia (E.H.B., J.S., M.F.); Department of Epidemiology & Preventative Medicine, Monash University, Melbourne, Australia (A.M.T.); Green Lane Cardiovascular Research Unit, Auckland City Hospital, Auckland, New Zealand (H.D.W.); Department of Medicine, University of Queensland, Brisbane, Australia (D.M.C.)
| | - Elizabeth H. Barnes
- From the Lipoprotein and Atherosclerosis Laboratory, Baker IDI Heart & Diabetes Institute, Melbourne, Australia (P.J.N.); NHMRC Clinical Trials Centre, University of Sydney, Sydney, Australia (E.H.B., J.S., M.F.); Department of Epidemiology & Preventative Medicine, Monash University, Melbourne, Australia (A.M.T.); Green Lane Cardiovascular Research Unit, Auckland City Hospital, Auckland, New Zealand (H.D.W.); Department of Medicine, University of Queensland, Brisbane, Australia (D.M.C.)
| | - Andrew M. Tonkin
- From the Lipoprotein and Atherosclerosis Laboratory, Baker IDI Heart & Diabetes Institute, Melbourne, Australia (P.J.N.); NHMRC Clinical Trials Centre, University of Sydney, Sydney, Australia (E.H.B., J.S., M.F.); Department of Epidemiology & Preventative Medicine, Monash University, Melbourne, Australia (A.M.T.); Green Lane Cardiovascular Research Unit, Auckland City Hospital, Auckland, New Zealand (H.D.W.); Department of Medicine, University of Queensland, Brisbane, Australia (D.M.C.)
| | - John Simes
- From the Lipoprotein and Atherosclerosis Laboratory, Baker IDI Heart & Diabetes Institute, Melbourne, Australia (P.J.N.); NHMRC Clinical Trials Centre, University of Sydney, Sydney, Australia (E.H.B., J.S., M.F.); Department of Epidemiology & Preventative Medicine, Monash University, Melbourne, Australia (A.M.T.); Green Lane Cardiovascular Research Unit, Auckland City Hospital, Auckland, New Zealand (H.D.W.); Department of Medicine, University of Queensland, Brisbane, Australia (D.M.C.)
| | - Marion Fournier
- From the Lipoprotein and Atherosclerosis Laboratory, Baker IDI Heart & Diabetes Institute, Melbourne, Australia (P.J.N.); NHMRC Clinical Trials Centre, University of Sydney, Sydney, Australia (E.H.B., J.S., M.F.); Department of Epidemiology & Preventative Medicine, Monash University, Melbourne, Australia (A.M.T.); Green Lane Cardiovascular Research Unit, Auckland City Hospital, Auckland, New Zealand (H.D.W.); Department of Medicine, University of Queensland, Brisbane, Australia (D.M.C.)
| | - Harvey D. White
- From the Lipoprotein and Atherosclerosis Laboratory, Baker IDI Heart & Diabetes Institute, Melbourne, Australia (P.J.N.); NHMRC Clinical Trials Centre, University of Sydney, Sydney, Australia (E.H.B., J.S., M.F.); Department of Epidemiology & Preventative Medicine, Monash University, Melbourne, Australia (A.M.T.); Green Lane Cardiovascular Research Unit, Auckland City Hospital, Auckland, New Zealand (H.D.W.); Department of Medicine, University of Queensland, Brisbane, Australia (D.M.C.)
| | - David M. Colquhoun
- From the Lipoprotein and Atherosclerosis Laboratory, Baker IDI Heart & Diabetes Institute, Melbourne, Australia (P.J.N.); NHMRC Clinical Trials Centre, University of Sydney, Sydney, Australia (E.H.B., J.S., M.F.); Department of Epidemiology & Preventative Medicine, Monash University, Melbourne, Australia (A.M.T.); Green Lane Cardiovascular Research Unit, Auckland City Hospital, Auckland, New Zealand (H.D.W.); Department of Medicine, University of Queensland, Brisbane, Australia (D.M.C.)
| | - Stefan Blankenberg
- From the Lipoprotein and Atherosclerosis Laboratory, Baker IDI Heart & Diabetes Institute, Melbourne, Australia (P.J.N.); NHMRC Clinical Trials Centre, University of Sydney, Sydney, Australia (E.H.B., J.S., M.F.); Department of Epidemiology & Preventative Medicine, Monash University, Melbourne, Australia (A.M.T.); Green Lane Cardiovascular Research Unit, Auckland City Hospital, Auckland, New Zealand (H.D.W.); Department of Medicine, University of Queensland, Brisbane, Australia (D.M.C.)
| | - David R. Sullivan
- From the Lipoprotein and Atherosclerosis Laboratory, Baker IDI Heart & Diabetes Institute, Melbourne, Australia (P.J.N.); NHMRC Clinical Trials Centre, University of Sydney, Sydney, Australia (E.H.B., J.S., M.F.); Department of Epidemiology & Preventative Medicine, Monash University, Melbourne, Australia (A.M.T.); Green Lane Cardiovascular Research Unit, Auckland City Hospital, Auckland, New Zealand (H.D.W.); Department of Medicine, University of Queensland, Brisbane, Australia (D.M.C.)
| |
Collapse
|
60
|
Jacobson TA. Lipoprotein(a), cardiovascular disease, and contemporary management. Mayo Clin Proc 2013; 88:1294-311. [PMID: 24182706 DOI: 10.1016/j.mayocp.2013.09.003] [Citation(s) in RCA: 80] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/10/2013] [Revised: 09/16/2013] [Accepted: 09/17/2013] [Indexed: 12/29/2022]
Abstract
Elevated lipoprotein(a) (Lp[a]) is a causal genetic risk factor for cardiovascular disease. To determine if current evidence supports both screening and treatment for elevated Lp(a) in high-risk patients, an English-language search of PubMed and MEDLINE was conducted. In population studies, there is a continuous association between Lp(a) concentrations and cardiovascular risk, with synergistic effects when low-density lipoprotein (LDL) is also elevated. Candidates for Lp(a) screening include patients with a personal or family history of premature cardiovascular disease, familial hypercholesterolemia, recurrent cardiovascular events, or inadequate LDL cholesterol (LDL-C) responses to statins. Given the comparative strength of clinical evidence, reducing LDL-C to the lowest attainable value with a high-potency statin should be the primary focus of lipid-modifying therapies. If the Lp(a) level is 30 mg/dL or higher in a patient who has the aforementioned characteristics plus residual LDL-C elevations (≥70-100 mg/dL) despite maximum-potency statins or combination statin therapy, the clinician may consider adding niacin (up to 2 g/d). If, after these interventions, the patient has progressive coronary heart disease (CHD) or LDL-C levels of 160-200 mg/dL or higher, LDL apheresis should be contemplated. Although Lp(a) is a major causal risk factor for CHD, no currently available controlled studies have suggested that lowering it through either pharmacotherapy or LDL apheresis specifically and significantly reduces coronary risk. Further research is needed to (1) optimize management in order to reduce CHD risk associated with elevated Lp(a) and (2) determine what other intermediate- or high-risk groups might benefit from Lp(a) screening.
Collapse
Affiliation(s)
- Terry A Jacobson
- Office of Health Promotion and Disease Prevention, Department of Medicine, Emory University School of Medicine, Atlanta, GA.
| |
Collapse
|
61
|
Todoric J, Handisurya A, Leitner K, Harreiter J, Hoermann G, Kautzky-Willer A. Lipoprotein(a) is not related to markers of insulin resistance in pregnancy. Cardiovasc Diabetol 2013; 12:138. [PMID: 24083682 PMCID: PMC3849879 DOI: 10.1186/1475-2840-12-138] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2013] [Accepted: 09/30/2013] [Indexed: 01/13/2023] Open
Abstract
BACKGROUND Dyslipidemia, a major risk factor for cardiovascular disease is a common finding in patients with type 2 diabetes and among women with gestational diabetes. Elevated levels of lipoprotein(a) [Lp(a)] are linked to increased risk of cardiovascular disease. However, its relationship with insulin resistance, type 2 diabetes and gestational diabetes is controversial and unproven. Here we aimed to clarify whether Lp(a) levels are associated with insulin sensitivity in pregnancy. METHODS Sixty-four women with gestational diabetes and 165 with normal glucose tolerance were enrolled in the study. Fasting Lp(a) serum levels were measured in all women at 24-28 weeks of gestation. RESULTS In pregnancy, there was no significant difference in serum Lp(a) concentrations between the two groups. Its level did not correlate with markers of insulin resistance (HOMA-IR), insulin sensitivity (HOMA-S%), pancreatic beta-cell function (HOMA-B%) and insulin sensitivity in dynamic conditions (OGIS). In addition, fasting glucose and insulin levels and those throughout an oral glucose tolerance test were independent of Lp(a) concentrations in our study group. CONCLUSIONS Lp(a) levels in pregnant women do not differ with respect to the presence or absence of gestational diabetes. Although influenced by some components of the lipid profile, such as triglycerides and HDL-C, insulin resistance in pregnancy is not affected by Lp(a).
Collapse
Affiliation(s)
- Jelena Todoric
- Laboratory of Gene Regulation and Signal Transduction, Departments of Pharmacology and Pathology, School of Medicine, University of California San Diego, 9500 Gilman Dr, La Jolla, CA 92093-0636, USA
- Department of Laboratory Medicine, Medical University of Vienna, Waehringer Guertel 18-20, 1090 Vienna, Austria
| | - Ammon Handisurya
- Department of Internal Medicine III, Division of Nephrology and Dialysis, Medical University of Vienna, Waehringer Guertel 18-20, 1090 Vienna, Austria
| | - Karoline Leitner
- Department of Internal Medicine III, Division of Endocrinology and Metabolism, Medical University of Vienna, Waehringer Guertel 18-20, 1090 Vienna, Austria
| | - Juergen Harreiter
- Department of Internal Medicine III, Division of Endocrinology and Metabolism, Medical University of Vienna, Waehringer Guertel 18-20, 1090 Vienna, Austria
| | - Gregor Hoermann
- Department of Laboratory Medicine, Medical University of Vienna, Waehringer Guertel 18-20, 1090 Vienna, Austria
| | - Alexandra Kautzky-Willer
- Department of Internal Medicine III, Division of Endocrinology and Metabolism, Medical University of Vienna, Waehringer Guertel 18-20, 1090 Vienna, Austria
| |
Collapse
|
62
|
Prognostic significance of elevated lipoprotein(a) in coronary artery revascularization patients. Int J Cardiol 2013; 167:1990-4. [DOI: 10.1016/j.ijcard.2012.05.007] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/27/2011] [Revised: 04/06/2012] [Accepted: 05/04/2012] [Indexed: 11/21/2022]
|
63
|
Li ZG, Li G, Zhou YL, Chen ZJ, Yang JQ, Zhang Y, Sun S, Zhong SL. Lack of association between lipoprotein(a) genetic variants and subsequent cardiovascular events in Chinese Han patients with coronary artery disease after percutaneous coronary intervention. Lipids Health Dis 2013; 12:127. [PMID: 23978127 PMCID: PMC3766040 DOI: 10.1186/1476-511x-12-127] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2012] [Accepted: 08/21/2013] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Elevated lipoprotein(a) [Lp(a)] levels predict cardiovascular events incidence in patients with coronary artery disease (CAD). Genetic variants in the rs3798220, rs10455872 and rs6415084 single-nucleotide polymorphisms (SNPs) in the Lp(a) gene (LPA) correlate with elevated Lp(a) levels, but whether these SNPs have prognostic value for CAD patients is unknown. The present study evaluated the association of LPA SNPs with incidence of subsequent cardiovascular events in CAD patients after percutaneous coronary intervention (PCI). METHODS TaqMan SNP genotyping assays were performed to detect the rs6415084, rs3798220 and rs10455872 genotypes in 517 Chinese Han patients with CAD after PCI. We later assessed whether there was an association of these SNPs with incidence of major adverse cardiovascular events (MACE: cardiac death, nonfatal myocardial infarction, ischemic stroke and coronary revascularization). Serum lipid profiles were also determined using biochemical methods. RESULTS Only the rs6415084 variant allele was associated with higher Lp(a) levels [41.3 (20.8, 74.6) vs. 18.6 (10.3, 40.9) mg/dl, p < 0.001]. During a 2-year follow-up period, 102 patients suffered MACE, and Cox regression analysis demonstrated that elevated Lp(a) (≥30 mg/dl) levels correlated with increased MACE (adjusted HR, 1.69; 95% CI 1.13-2.53), but there was no association between LPA genetic variants (rs6415084 and rs3798220) and MACE incidence (p > 0.05). CONCLUSIONS Our data did not support a relationship between genetic LPA variants (rs6415084 and rs3798220) and subsequent cardiovascular events after PCI in Chinese Han CAD patients.
Collapse
Affiliation(s)
- Zhi-Gen Li
- Department of Cardiology, Guangdong Cardiovascular Institute, Guangdong Academy of Medical Sciences, Guangdong General Hospital, 96 Dongchuan Road, Guangzhou 510007, China.
| | | | | | | | | | | | | | | |
Collapse
|
64
|
Hoover-Plow J, Huang M. Lipoprotein(a) metabolism: potential sites for therapeutic targets. Metabolism 2013; 62:479-91. [PMID: 23040268 PMCID: PMC3547132 DOI: 10.1016/j.metabol.2012.07.024] [Citation(s) in RCA: 112] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/04/2012] [Revised: 07/03/2012] [Accepted: 07/11/2012] [Indexed: 11/20/2022]
Abstract
Lipoprotein(a) [Lp(a)] resembles low-density lipoprotein (LDL), with an LDL lipid core and apolipoprotein B (apoB), but contains a unique apolipoprotein, apo(a). Elevated Lp(a) is an independent risk factor for coronary and peripheral vascular diseases. The size and concentration of plasma Lp(a) are related to the synthetic rate, not the catabolic rate, and are highly variable with small isoforms associated with high concentrations and pathogenic risk. Apo(a) is synthesized in the liver, although assembly of apo(a) and LDL may occur in the hepatocytes or plasma. While the uptake and clearance site of Lp(a) is poorly delineated, the kidney is the site of apo(a) fragment excretion. The structure of apo(a) has high homology to plasminogen, the zymogen for plasmin and the primary clot lysis enzyme. Apo(a) interferes with plasminogen binding to C-terminal lysines of cell surface and extracellular matrix proteins. Lp(a) and apo(a) inhibit fibrinolysis and accumulate in the vascular wall in atherosclerotic lesions. The pathogenic role of Lp(a) is not known. Small isoforms and high concentrations of Lp(a) are found in healthy octogenarians that suggest Lp(a) may also have a physiological role. Studies of Lp(a) function have been limited since it is not found in commonly studied small mammals. An important aspect of Lp(a) metabolism is the modification of circulating Lp(a), which has the potential to alter the functions of Lp(a). There are no therapeutic drugs that selectively target elevated Lp(a), but a number of possible agents are being considered. Recently, new modifiers of apo(a) synthesis have been identified. This review reports the regulation of Lp(a) metabolism and potential sites for therapeutic targets.
Collapse
Affiliation(s)
- Jane Hoover-Plow
- J. J. Jacobs Center for Thrombosis and Vascular Biology, Department of Cardiovascular Medicine, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44139, USA.
| | | |
Collapse
|
65
|
Li Y, Luke MM, Shiffman D, Devlin JJ. Genetic variants in the apolipoprotein(a) gene and coronary heart disease. ACTA ACUST UNITED AC 2012; 4:565-73. [PMID: 22010162 DOI: 10.1161/circgenetics.111.959601] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
|
66
|
Morgan TM, House JA, Cresci S, Jones P, Allayee H, Hazen SL, Patel Y, Patel RS, Eapen DJ, Waddy SP, Quyyumi AA, Kleber ME, März W, Winkelmann BR, Boehm BO, Krumholz HM, Spertus JA. Investigation of 95 variants identified in a genome-wide study for association with mortality after acute coronary syndrome. BMC MEDICAL GENETICS 2011; 12:127. [PMID: 21957892 PMCID: PMC3190329 DOI: 10.1186/1471-2350-12-127] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/27/2010] [Accepted: 09/29/2011] [Indexed: 11/13/2022]
Abstract
Background Genome-wide association studies (GWAS) have identified new candidate genes for the occurrence of acute coronary syndrome (ACS), but possible effects of such genes on survival following ACS have yet to be investigated. Methods We examined 95 polymorphisms in 69 distinct gene regions identified in a GWAS for premature myocardial infarction for their association with post-ACS mortality among 811 whites recruited from university-affiliated hospitals in Kansas City, Missouri. We then sought replication of a positive genetic association in a large, racially diverse cohort of myocardial infarction patients (N = 2284) using Kaplan-Meier survival analyses and Cox regression to adjust for relevant covariates. Finally, we investigated the apparent association further in 6086 additional coronary artery disease patients. Results After Cox adjustment for other ACS risk factors, of 95 SNPs tested in 811 whites only the association with the rs6922269 in MTHFD1L was statistically significant, with a 2.6-fold mortality hazard (P = 0.007). The recessive A/A genotype was of borderline significance in an age- and race-adjusted analysis of the entire combined cohort (N = 3095; P = 0.052), but this finding was not confirmed in independent cohorts (N = 6086). Conclusions We found no support for the hypothesis that the GWAS-identified variants in this study substantially alter the probability of post-ACS survival. Large-scale, collaborative, genome-wide studies may be required in order to detect genetic variants that are robustly associated with survival in patients with coronary artery disease.
Collapse
Affiliation(s)
- Thomas M Morgan
- Department of Pediatrics, Vanderbilt University School of Medicine, Nashville, TN, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
67
|
Chennamsetty I, Claudel T, Kostner KM, Baghdasaryan A, Kratky D, Levak-Frank S, Frank S, Gonzalez FJ, Trauner M, Kostner GM. Farnesoid X receptor represses hepatic human APOA gene expression. J Clin Invest 2011; 121:3724-34. [PMID: 21804189 DOI: 10.1172/jci45277] [Citation(s) in RCA: 81] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2011] [Accepted: 06/01/2011] [Indexed: 12/31/2022] Open
Abstract
High plasma concentrations of lipoprotein(a) [Lp(a), which is encoded by the APOA gene] increase an individual's risk of developing diseases, such as coronary artery diseases, restenosis, and stroke. Unfortunately, increased Lp(a) levels are minimally influenced by dietary changes or drug treatment. Further, the development of Lp(a)-specific medications has been hampered by limited knowledge of Lp(a) metabolism. In this study, we identified patients suffering from biliary obstructions with very low plasma Lp(a) concentrations that rise substantially after surgical intervention. Consistent with this, common bile duct ligation in mice transgenic for human APOA (tg-APOA mice) lowered plasma concentrations and hepatic expression of APOA. To test whether farnesoid X receptor (FXR), which is activated by bile acids, was responsible for the low plasma Lp(a) levels in cholestatic patients and mice, we treated tg-APOA and tg-APOA/Fxr-/- mice with cholic acid. FXR activation markedly reduced plasma concentrations and hepatic expression of human APOA in tg-APOA mice but not in tg-APOA/Fxr-/- mice. Incubation of primary hepatocytes from tg-APOA mice with bile acids dose dependently downregulated APOA expression. Further analysis determined that the direct repeat 1 element between nucleotides -826 and -814 of the APOA promoter functioned as a negative FXR response element. This motif is also bound by hepatocyte nuclear factor 4α (HNF4α), which promotes APOA transcription, and FXR was shown to compete with HNF4α for binding to this motif. These findings may have important implications in the development of Lp(a)-lowering medications.
Collapse
Affiliation(s)
- Indumathi Chennamsetty
- Institute of Molecular Biology and Biochemistry, Center of Molecular Medicine, Medical University of Graz, Graz, Austria
| | | | | | | | | | | | | | | | | | | |
Collapse
|
68
|
Nenseter MS, Lindvig HW, Ueland T, Langslet G, Ose L, Holven KB, Retterstøl K. Lipoprotein(a) levels in coronary heart disease-susceptible and -resistant patients with familial hypercholesterolemia. Atherosclerosis 2011; 216:426-32. [PMID: 21376325 DOI: 10.1016/j.atherosclerosis.2011.02.007] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2010] [Revised: 02/02/2011] [Accepted: 02/03/2011] [Indexed: 10/18/2022]
|
69
|
Hartiala J, Li D, Conti DV, Vikman S, Patel Y, Wilson Tang WH, Brennan ML, Newman JW, Stephensen CB, Armstrong P, Hazen SL, Allayee H. Genetic contribution of the leukotriene pathway to coronary artery disease. Hum Genet 2011; 129:617-27. [PMID: 21293878 PMCID: PMC3092945 DOI: 10.1007/s00439-011-0963-3] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2010] [Accepted: 01/27/2011] [Indexed: 12/25/2022]
Abstract
We evaluated the genetic contribution of the leukotriene (LT) pathway to risk of coronary artery disease (CAD) in 4,512 Caucasian and African American subjects ascertained through elective cardiac evaluation. Of the three previously associated variants, the shorter "3" and "4" alleles of a promoter repeat polymorphism in ALOX5 increased risk of CAD in African Americans (OR = 1.4, 95% CI 1.0-1.9; p = 0.04), whereas a haplotype of LTA4H (HapK) was associated with CAD in Caucasians (OR = 1.2, 95% CI 1.01-1.4; p = 0.03). In Caucasians, first-stage analysis of 254 haplotype-tagging SNPs in 15 LT pathway genes with follow-up of 19 variants in stage 2 revealed an LTA4H SNP (rs2540477) that increased risk of CAD (OR = 1.2, 95% CI 1.1-1.5; p = 0.003) and a PLA2G4A SNP (rs12746200) that decreased risk of CAD (OR = 0.7, 95% CI 0.6-0.9; p = 0.0007). The PLA2G4A rs12746200 variant also decreased risk of experiencing a major adverse cardiac event (MACE = myocardial infarction, stroke, or death) over 3 years of follow-up (HR = 0.7, 95% CI 0.5-0.9; p = 0.01), consistent with its cardioprotective effect. Functional experiments demonstrated that stimulated monocytes from carriers of LTA4H variants HapK or rs2540477 had 50% (p = 0.002) and 33% (p = 0.03) higher LTB(4) production, respectively, compared to non-carriers. These ex vivo results are consistent with LTB(4) being the direct product of the reaction catalyzed by LTA4H and its role in promoting monocyte chemotaxis to sites of inflammation, including the artery wall of atherosclerotic lesions. Taken together, this study provides additional evidence that functional genetic variation of the LT pathway can mediate atherogenic processes and the risk of CAD in humans.
Collapse
Affiliation(s)
- Jaana Hartiala
- Department of Preventive Medicine, USC Keck School of Medicine, Los Angeles, CA 90033 USA
- Institute for Genetic Medicine, USC Keck School of Medicine, 2250 Alcazar Street, CSC 206, Los Angeles, CA 90033 USA
| | - Dalin Li
- Department of Preventive Medicine, USC Keck School of Medicine, Los Angeles, CA 90033 USA
| | - David V. Conti
- Department of Preventive Medicine, USC Keck School of Medicine, Los Angeles, CA 90033 USA
| | - Susanna Vikman
- Department of Preventive Medicine, USC Keck School of Medicine, Los Angeles, CA 90033 USA
- Institute for Genetic Medicine, USC Keck School of Medicine, 2250 Alcazar Street, CSC 206, Los Angeles, CA 90033 USA
| | - Yesha Patel
- Department of Preventive Medicine, USC Keck School of Medicine, Los Angeles, CA 90033 USA
- Institute for Genetic Medicine, USC Keck School of Medicine, 2250 Alcazar Street, CSC 206, Los Angeles, CA 90033 USA
| | - W. H. Wilson Tang
- Department of Cardiovascular Medicine, Cleveland Clinic, Cleveland, OH 44195 USA
| | - Marie-Louise Brennan
- Department of Cardiovascular Medicine, Cleveland Clinic, Cleveland, OH 44195 USA
- Department of Cell Biology, Cleveland Clinic, Cleveland, OH 44195 USA
- Center for Cardiovascular Diagnostics and Prevention, Cleveland Clinic, Cleveland, OH 44195 USA
| | - John W. Newman
- USDA Western Human Nutrition Research Center, University of California Davis, Davis, CA 95616 USA
| | - Charles B. Stephensen
- USDA Western Human Nutrition Research Center, University of California Davis, Davis, CA 95616 USA
- Program in International and Community Nutrition, Department of Nutrition, University of California Davis, Davis, CA 95616 USA
| | - Patrice Armstrong
- USDA Western Human Nutrition Research Center, University of California Davis, Davis, CA 95616 USA
- Program in International and Community Nutrition, Department of Nutrition, University of California Davis, Davis, CA 95616 USA
| | - Stanley L. Hazen
- Department of Cardiovascular Medicine, Cleveland Clinic, Cleveland, OH 44195 USA
- Department of Cell Biology, Cleveland Clinic, Cleveland, OH 44195 USA
- Center for Cardiovascular Diagnostics and Prevention, Cleveland Clinic, Cleveland, OH 44195 USA
| | - Hooman Allayee
- Department of Preventive Medicine, USC Keck School of Medicine, Los Angeles, CA 90033 USA
- Institute for Genetic Medicine, USC Keck School of Medicine, 2250 Alcazar Street, CSC 206, Los Angeles, CA 90033 USA
| |
Collapse
|
70
|
Wang Z, Klipfell E, Bennett BJ, Koeth R, Levison BS, Dugar B, Feldstein AE, Britt EB, Fu X, Chung YM, Wu Y, Schauer P, Smith JD, Allayee H, Tang WHW, DiDonato JA, Lusis AJ, Hazen SL. Gut flora metabolism of phosphatidylcholine promotes cardiovascular disease. Nature 2011; 472:57-63. [PMID: 21475195 PMCID: PMC3086762 DOI: 10.1038/nature09922] [Citation(s) in RCA: 3982] [Impact Index Per Article: 284.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2009] [Accepted: 02/09/2011] [Indexed: 02/06/2023]
Abstract
Metabolomics studies hold promise for discovery of pathways linked to disease processes. Cardiovascular disease (CVD) represents the leading cause of death and morbidity worldwide. A metabolomics approach was used to generate unbiased small molecule metabolic profiles in plasma that predict risk for CVD. Three metabolites of the dietary lipid phosphatidylcholine, namely choline, trimethylamine N-oxide (TMAO), and betaine, were identified and then shown to predict risk for CVD in an independent large clinical cohort. Dietary supplementation of mice with choline, TMAO or betaine promoted up-regulation of multiple macrophage scavenger receptors linked to atherosclerosis, and supplementation with choline or TMAO promoted atherosclerosis. Studies using germ-free mice confirmed a critical role for dietary choline and gut flora in TMAO production, augmented macrophage cholesterol accumulation and foam cell formation. Suppression of intestinal microflora in atherosclerosis-prone mice inhibited dietary choline-enhanced atherosclerosis. Genetic variations controlling expression of flavin monooxygenases (FMOs), an enzymatic source of TMAO, segregated with atherosclerosis in hyperlipidemic mice. Discovery of a relationship between gut flora-dependent metabolism of dietary phosphatidylcholine and CVD pathogenesis provides opportunities for development of both novel diagnostic tests and therapeutic approaches for atherosclerotic heart disease.
Collapse
Affiliation(s)
- Zeneng Wang
- Department of Cell Biology, Cleveland Clinic, Cleveland, Ohio 44195, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|