1
|
Lian PA, Zhu WQ, Zhao WX, Huang PP, Ran JL, Tang YX, Huang XS, Li R. Lipoprotein(a) in atherosclerotic cardiovascular disease and proprotein convertase subtilisin/kexin-type 9 inhibitors. Clin Chim Acta 2024; 565:119982. [PMID: 39366516 DOI: 10.1016/j.cca.2024.119982] [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: 02/08/2024] [Revised: 09/28/2024] [Accepted: 09/29/2024] [Indexed: 10/06/2024]
Abstract
High plasma lipoprotein(a) (Lp(a)) levels increase the cardiovascular risk in populations with atherosclerotic cardiovascular disease (ASCVD). Apolipoprotein (a) [apo(a)], a unique protein component of Lp(a), plays an important role in the pathogenesis of atherosclerosis. Statins, the primary medication in managing ASCVD, lower low-density lipoprotein cholesterol (LDL-C) but concurrently elevate plasma Lp(a) levels, contributing to an increased residual cardiovascular risk. In turn, proprotein convertase subtilisin/kexin-type 9 (PCSK9) inhibitors, a novel class of LDL-C lowering drugs, effectively reduce plasma Lp(a) levels, which is believed to decrease residual cardiovascular risk. However, the mechanism by which PCSK9 inhibitors reduce Lp(a) levels remains unknown. In addition, there are some clinical limitations of PCSK9 inhibitors. Here, we systematically review the past, present, and prospects of studies pertaining to Lp(a), PCSK9 inhibitors, and ASCVD.
Collapse
Affiliation(s)
- Ping-An Lian
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Wen-Qiang Zhu
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Wei-Xin Zhao
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Piao-Piao Huang
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Juan-Li Ran
- Department of Stomatology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Ya-Xin Tang
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Xian-Sheng Huang
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China; Department of Cardiovascular Medicine, Guilin Hospital of The Second Xiangya Hospital, Central South University, Guilin, China
| | - Rong Li
- Department of Stomatology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China; Department of Stomatology, Guilin Hospital of The Second Xiangya Hospital, Central South University, Guilin, China.
| |
Collapse
|
2
|
Kris-Etherton PM, Riley TM, Petersen KS. Dietary modulation of Lp(a): more questions than answers. J Lipid Res 2024; 65:100592. [PMID: 38996919 PMCID: PMC11345285 DOI: 10.1016/j.jlr.2024.100592] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/14/2024] Open
Affiliation(s)
- Penny M Kris-Etherton
- Department of Nutritional Sciences, Pennsylvania State University, University Park, PA, USA
| | - Terrence M Riley
- Nutritional Physiology Lab, Pennington Biomedical Research Center, Baton Rouge, LA, USA
| | - Kristina S Petersen
- Department of Nutritional Sciences, Pennsylvania State University, University Park, PA, USA.
| |
Collapse
|
3
|
Akyol O, Yang CY, Woodside DG, Chiang HH, Chen CH, Gotto AM. Comparative Analysis of Atherogenic Lipoproteins L5 and Lp(a) in Atherosclerotic Cardiovascular Disease. Curr Atheroscler Rep 2024; 26:317-329. [PMID: 38753254 PMCID: PMC11192678 DOI: 10.1007/s11883-024-01209-3] [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] [Accepted: 05/05/2024] [Indexed: 06/22/2024]
Abstract
PURPOSE OF REVIEW Low-density lipoprotein (LDL) poses a risk for atherosclerotic cardiovascular disease (ASCVD). As LDL comprises various subtypes differing in charge, density, and size, understanding their specific impact on ASCVD is crucial. Two highly atherogenic LDL subtypes-electronegative LDL (L5) and Lp(a)-induce vascular cell apoptosis and atherosclerotic changes independent of plasma cholesterol levels, and their mechanisms warrant further investigation. Here, we have compared the roles of L5 and Lp(a) in the development of ASCVD. RECENT FINDINGS Lp(a) tends to accumulate in artery walls, promoting plaque formation and potentially triggering atherosclerosis progression through prothrombotic or antifibrinolytic effects. High Lp(a) levels correlate with calcific aortic stenosis and atherothrombosis risk. L5 can induce endothelial cell apoptosis and increase vascular permeability, inflammation, and atherogenesis, playing a key role in initiating atherosclerosis. Elevated L5 levels in certain high-risk populations may serve as a distinctive predictor of ASCVD. L5 and Lp(a) are both atherogenic lipoproteins contributing to ASCVD through distinct mechanisms. Lp(a) has garnered attention, but equal consideration should be given to L5.
Collapse
Affiliation(s)
- Omer Akyol
- Molecular Cardiology Research Laboratories, Vascular and Medicinal Research, The Texas Heart Institute, Houston, Texas, 77030, USA
| | - Chao-Yuh Yang
- Department of Medicine, Baylor College of Medicine, One Baylor Plaza, Houston, Texas, 77030, USA
| | - Darren G Woodside
- Molecular Cardiology Research Laboratories, The Texas Heart Institute, Houston, TX, 77030, USA
| | - Huan-Hsing Chiang
- Molecular Cardiology Research Laboratories, Vascular and Medicinal Research, The Texas Heart Institute, Houston, Texas, 77030, USA
| | - Chu-Huang Chen
- Molecular Cardiology Research Laboratories, Vascular and Medicinal Research, The Texas Heart Institute, Houston, Texas, 77030, USA.
| | | |
Collapse
|
4
|
Khan TG, Cunha JB, Raut C, Burroughs M, Goonewardena SN, Smrcka AV, Speliotes EK, Emmer BT. Functional interrogation of cellular Lp(a) uptake by genome-scale CRISPR screening. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.11.593568. [PMID: 38766193 PMCID: PMC11100788 DOI: 10.1101/2024.05.11.593568] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/22/2024]
Abstract
An elevated level of lipoprotein(a), or Lp(a), in the bloodstream has been causally linked to the development of atherosclerotic cardiovascular disease and calcific aortic valve stenosis. Steady state levels of circulating lipoproteins are modulated by their rate of clearance, but the identity of the Lp(a) uptake receptor(s) has been controversial. In this study, we performed a genome-scale CRISPR screen to functionally interrogate all potential Lp(a) uptake regulators in HuH7 cells. Strikingly, the top positive and negative regulators of Lp(a) uptake in our screen were LDLR and MYLIP, encoding the LDL receptor and its ubiquitin ligase IDOL, respectively. We also found a significant correlation for other genes with established roles in LDLR regulation. No other gene products, including those previously proposed as Lp(a) receptors, exhibited a significant effect on Lp(a) uptake in our screen. We validated the functional influence of LDLR expression on HuH7 Lp(a) uptake, confirmed in vitro binding between the LDLR extracellular domain and purified Lp(a), and detected an association between loss-of-function LDLR variants and increased circulating Lp(a) levels in the UK Biobank cohort. Together, our findings support a central role for the LDL receptor in mediating Lp(a) uptake by hepatocytes.
Collapse
Affiliation(s)
- Taslima G. Khan
- Program in Chemical Biology, University of Michigan, Ann Arbor MI
| | - Juliana Bragazzi Cunha
- Division of Hospital Medicine, Department of Internal Medicine, University of Michigan, Ann Arbor MI
| | - Chinmay Raut
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, University of Michigan, Ann Arbor MI
- Department of Computational Medicine and Bioinformatics, University of Michigan Medical School, Ann Arbor MI
| | | | - Sascha N. Goonewardena
- Division of Cardiovascular Medicine, Department of Internal Medicine, University of Michigan, Ann Arbor MI
- Frankel Cardiovascular Center, University of Michigan, Ann Arbor MI
| | - Alan V. Smrcka
- Department of Pharmacology, University of Michigan, Ann Arbor MI
| | - Elizabeth K. Speliotes
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, University of Michigan, Ann Arbor MI
- Department of Computational Medicine and Bioinformatics, University of Michigan Medical School, Ann Arbor MI
| | - Brian T. Emmer
- Division of Hospital Medicine, Department of Internal Medicine, University of Michigan, Ann Arbor MI
- Frankel Cardiovascular Center, University of Michigan, Ann Arbor MI
| |
Collapse
|
5
|
Ugovšek S, Rehberger Likozar A, Levstek T, Trebušak Podkrajšek K, Zupan J, Šebeštjen M. Haplotype of the Lipoprotein(a) Gene Variants rs10455872 and rs3798220 Is Associated with Parameters of Coagulation, Fibrinolysis, and Inflammation in Patients after Myocardial Infarction and Highly Elevated Lipoprotein(a) Values. Int J Mol Sci 2024; 25:736. [PMID: 38255810 PMCID: PMC10815733 DOI: 10.3390/ijms25020736] [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: 12/05/2023] [Revised: 12/20/2023] [Accepted: 01/04/2024] [Indexed: 01/24/2024] Open
Abstract
Lipoprotein(a) (Lp(a)) is an independent risk factor for future coronary events. Variants rs10455872 and rs3798220 in the gene encoding Lp(a) are associated with an increased Lp(a) concentration and risk of coronary artery disease. We aimed to determine whether in high-risk coronary artery disease patients these two genetic variants and the kringle IV type 2 (KIV-2) repeats are associated with impairment of inflammatory and hemostatic parameters. Patients after myocardial infarction with elevated Lp(a) levels were included. Blood samples underwent biochemical and genetic analyses. In carriers of the AC haplotype, the concentrations of tumor necrosis factor (TNF)-α (4.46 vs. 3.91 ng/L, p = 0.046) and plasminogen activator inhibitor-1 (PAI-1) (p = 0.026) were significantly higher compared to non-carriers. The number of KIV-2 repeats was significantly associated with the concentration of high-sensitivity C-reactive protein (ρ = 0.251, p = 0.038) and overall fibrinolytic potential (r = -0.253, p = 0.038). In our patients, a direct association between the AC haplotype and both TNF-α and PAI-1 levels was observed. Our study shows that the number of KIV-2 repeats not only affects proatherosclerotic and proinflammatory effects of Lp(a) but is also associated with its antifibrinolytic properties.
Collapse
Affiliation(s)
- Sabina Ugovšek
- Division of Internal Medicine, University Medical Centre Ljubljana, Zaloška cesta 7, 1000 Ljubljana, Slovenia
- Faculty of Medicine, University of Ljubljana, Vrazov trg 2, 1000 Ljubljana, Slovenia
| | - Andreja Rehberger Likozar
- Department of Vascular Diseases, University Medical Centre Ljubljana, Zaloška cesta 7, 1000 Ljubljana, Slovenia;
| | - Tina Levstek
- Laboratory for Translational Medical Biochemistry, Institute of Biochemistry and Molecular Genetics, Faculty of Medicine, University of Ljubljana, Vrazov trg 2, 1000 Ljubljana, Slovenia; (T.L.); (K.T.P.)
- Clinical Institute for Special Laboratory Diagnostics, University Children’s Hospital, University Medical Centre Ljubljana, Vrazov trg 1, 1000 Ljubljana, Slovenia
| | - Katarina Trebušak Podkrajšek
- Laboratory for Translational Medical Biochemistry, Institute of Biochemistry and Molecular Genetics, Faculty of Medicine, University of Ljubljana, Vrazov trg 2, 1000 Ljubljana, Slovenia; (T.L.); (K.T.P.)
- Clinical Institute for Special Laboratory Diagnostics, University Children’s Hospital, University Medical Centre Ljubljana, Vrazov trg 1, 1000 Ljubljana, Slovenia
| | - Janja Zupan
- Faculty of Pharmacy, University of Ljubljana, Aškerčeva cesta 7, 1000 Ljubljana, Slovenia;
| | - Miran Šebeštjen
- Faculty of Medicine, University of Ljubljana, Vrazov trg 2, 1000 Ljubljana, Slovenia
- Department of Vascular Diseases, University Medical Centre Ljubljana, Zaloška cesta 7, 1000 Ljubljana, Slovenia;
- Department of Cardiology, University Medical Centre Ljubljana, Zaloška cesta 7, 1000 Ljubljana, Slovenia
| |
Collapse
|
6
|
The impact of the PCSK-9/VLDL-Receptor axis on inflammatory cell polarization. Cytokine 2023; 161:156077. [PMID: 36356495 DOI: 10.1016/j.cyto.2022.156077] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Revised: 09/26/2022] [Accepted: 10/14/2022] [Indexed: 11/09/2022]
Abstract
BACKGROUND Studies have shown that lipoproteins, such as LDL and VLDL, as well as its major protein component ApoE2 impact on macrophage polarization important in atherosclerosis. Proprotein convertase subtilisin/kexin 9 (PCSK9) is a key regulator of lipoprotein receptor expression. The present study investigated the effect of the VLDL/VLDL-receptor (VLDL-R) axis on mononuclear cell polarization, as well as the role of PCSK9 and PCSK9 inhibitors (PCSK9i) within this network. METHODS Human monocytic THP-1 cells and human monocyte-derived macrophages isolated from peripheral blood mononuclear cells (PBMC) were treated with either LPS/IFN-γ to induce a pro-inflammatory phenotype, or with IL-4/IL-13 to induce an anti-inflammatory phenotype. Cells were then subjected to further treatments by lipoproteins, PCSK9, PCSK9i and lipoprotein receptor blockers. RESULTS LPS/IFN-γ treatment promoted a pro-inflammatory state with an increased expression of pro-inflammatory mediators such as TNF-α, CD80 and IL-1β. VLDL co-treatment induced a switch of this pro-inflammatory phenotype to an anti-inflammatory phenotype. In pro-inflammatory cells, VLDL significantly decreased the expression of pro-inflammatory markers e.g., TNF-α, CD80, and IL-1β. These effects were eliminated by PCSK9 and restored by co-incubation with a specific anti-PCSK9 monoclonal antibody (PCSK9i). Migration assays demonstrated that pro-inflammatory cells displayed a significantly higher invasive capacity when compared to untreated cells or anti-inflammatory cells. Moreover, pro-inflammatory cell chemotaxis was significantly decreased by VLDL-mediated acquisition of the anti-inflammatory phenotype. PCSK9 significantly lessened this VLDL-mediated migration inhibition, which was reversed by the PCSK9i. CONCLUSION VLDL promotes mononuclear cell differentiation towards an anti-inflammatory phenotype. PCSK9, via its capacity to inhibit VLDL-R expression, reverses the VLDL-mediated anti-inflammatory action, thereby promoting a pro-inflammatory phenotype. Thus, PCSK9 targeting therapies may exert anti-inflammatory properties within the vessel wall.
Collapse
|
7
|
Chemello K, Chan DC, Lambert G, Watts GF. Recent advances in demystifying the metabolism of lipoprotein(a). Atherosclerosis 2022; 349:82-91. [DOI: 10.1016/j.atherosclerosis.2022.04.002] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Revised: 03/29/2022] [Accepted: 04/01/2022] [Indexed: 12/24/2022]
|
8
|
Clark JR, Gemin M, Youssef A, Marcovina SM, Prat A, Seidah NG, Hegele RA, Boffa MB, Koschinsky ML. Sortilin enhances secretion of apolipoprotein(a) through effects on apolipoprotein B secretion and promotes uptake of lipoprotein(a). J Lipid Res 2022; 63:100216. [PMID: 35469919 PMCID: PMC9131257 DOI: 10.1016/j.jlr.2022.100216] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Revised: 04/06/2022] [Accepted: 04/08/2022] [Indexed: 12/30/2022] Open
Abstract
Elevated plasma lipoprotein(a) (Lp(a)) is an independent, causal risk factor for atherosclerotic cardiovascular disease and calcific aortic valve stenosis. Lp(a) is formed in or on hepatocytes from successive noncovalent and covalent interactions between apo(a) and apoB, although the subcellular location of these interactions and the nature of the apoB-containing particle involved remain unclear. Sortilin, encoded by the SORT1 gene, modulates apoB secretion and LDL clearance. We used a HepG2 cell model to study the secretion kinetics of apo(a) and apoB. Overexpression of sortilin increased apo(a) secretion, while siRNA-mediated knockdown of sortilin expression correspondingly decreased apo(a) secretion. Sortilin binds LDL but not apo(a) or Lp(a), indicating that its effect on apo(a) secretion is likely indirect. Indeed, the effect was dependent on the ability of apo(a) to interact noncovalently with apoB. Overexpression of sortilin enhanced internalization of Lp(a), but not apo(a), by HepG2 cells, although neither sortilin knockdown in these cells or Sort1 deficiency in mice impacted Lp(a) uptake. We found several missense mutations in SORT1 in patients with extremely high Lp(a) levels; sortilin containing some of these mutations was more effective at promoting apo(a) secretion than WT sortilin, though no differences were found with respect to Lp(a) internalization. Our observations suggest that sortilin could play a role in determining plasma Lp(a) levels and corroborate in vivo human kinetic studies which imply that secretion of apo(a) and apoB are coupled, likely within the hepatocyte.
Collapse
Affiliation(s)
- Justin R Clark
- Department of Physiology & Pharmacology, Schulich School of Medicine & Dentistry, The University of Western Ontario, London, ON, Canada
| | - Matthew Gemin
- Department of Chemistry & Biochemistry, University of Windsor, Windsor, ON, Canada
| | - Amer Youssef
- Robarts Research Institute, Schulich School of Medicine & Dentistry, The University of Western Ontario, London, ON, Canada
| | | | - Annik Prat
- Institut de Recherches Cliniques de Montreal, Montréal, QC, Canada
| | - Nabil G Seidah
- Institut de Recherches Cliniques de Montreal, Montréal, QC, Canada
| | - Robert A Hegele
- Robarts Research Institute, Schulich School of Medicine & Dentistry, The University of Western Ontario, London, ON, Canada; Department of Biochemistry, Schulich School of Medicine & Dentistry, The University of Western Ontario, London, ON, Canada; Department of Medicine, Schulich School of Medicine & Dentistry, The University of Western Ontario, London, ON, Canada
| | - Michael B Boffa
- Robarts Research Institute, Schulich School of Medicine & Dentistry, The University of Western Ontario, London, ON, Canada; Department of Biochemistry, Schulich School of Medicine & Dentistry, The University of Western Ontario, London, ON, Canada
| | - Marlys L Koschinsky
- Department of Physiology & Pharmacology, Schulich School of Medicine & Dentistry, The University of Western Ontario, London, ON, Canada; Robarts Research Institute, Schulich School of Medicine & Dentistry, The University of Western Ontario, London, ON, Canada.
| |
Collapse
|
9
|
Kimura T, Akahori H, Tanaka T, Yanaka K, Yoshihara N, Miki K, Imanaka T, Asakura M, Ishihara M. Impact of lipoprotein (a) on long-term outcome after percutaneous coronary intervention in the era of new generation drug-eluting stents. J Cardiol 2022; 80:179-183. [DOI: 10.1016/j.jjcc.2022.03.004] [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: 12/25/2021] [Revised: 02/18/2022] [Accepted: 02/26/2022] [Indexed: 11/26/2022]
|
10
|
Rhainds D, Brodeur MR, Tardif JC. Lipoprotein (a): When to Measure and How to Treat? Curr Atheroscler Rep 2021; 23:51. [PMID: 34235598 DOI: 10.1007/s11883-021-00951-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/03/2021] [Indexed: 12/11/2022]
Abstract
PURPOSE OF REVIEW The purpose of this article is to review current evidence for lipoprotein (a) (Lp(a)) as a risk factor for multiple cardiovascular (CV) disease phenotypes, provide a rationale for Lp(a) lowering to reduce CV risk, identify therapies that lower Lp(a) levels that are available clinically and under investigation, and discuss future directions. RECENT FINDINGS Mendelian randomization and epidemiological studies have shown that elevated Lp(a) is an independent and causal risk factor for atherosclerosis and major CV events. Lp(a) is also associated with non-atherosclerotic endpoints such as venous thromboembolism and calcific aortic valve disease. It contributes to residual CV risk in patients receiving standard-of-care LDL-lowering therapy. Plasma Lp(a) levels present a skewed distribution towards higher values and vary widely between individuals and according to ethnic background due to genetic variants in the LPA gene, but remain relatively constant throughout a person's life. Thus, elevated Lp(a) (≥50 mg/dL) is a prevalent condition affecting >20% of the population but is still underdiagnosed. Treatment guidelines have begun to advocate measurement of Lp(a) to identify patients with very high levels that have a family history of premature CVD or elevated Lp(a). Lipoprotein apheresis (LA) efficiently lowers Lp(a) and was recently associated with a reduction of incident CV events. Statins have neutral or detrimental effects on Lp(a), while PCSK9 inhibitors significantly reduce its level by up to 30%. Specific lowering of Lp(a) with antisense oligonucleotides (ASO) shows good safety and strong efficacy with up to 90% reductions. The ongoing CV outcomes study Lp(a)HORIZON will provide a first answer as to whether selective Lp(a) lowering with ASO reduces the risk of major CV events. Given the recently established association between Lp(a) level and CV risk, guidelines now recommend Lp(a) measurement in specific clinical conditions. Accordingly, Lp(a) is a current target for drug development to reduce CV risk in patients with elevated levels, and lowering Lp(a) with ASO represents a promising avenue.
Collapse
Affiliation(s)
- David Rhainds
- Montreal Heart Institute Research Center, 5000 Belanger Street, Montréal, Canada
| | - Mathieu R Brodeur
- Montreal Heart Institute Research Center, 5000 Belanger Street, Montréal, Canada
| | - Jean-Claude Tardif
- Montreal Heart Institute Research Center, 5000 Belanger Street, Montréal, Canada. .,Faculty of Medicine, Université de Montréal, Montréal, Canada.
| |
Collapse
|
11
|
Mineo C. Lipoprotein receptor signalling in atherosclerosis. Cardiovasc Res 2021; 116:1254-1274. [PMID: 31834409 DOI: 10.1093/cvr/cvz338] [Citation(s) in RCA: 87] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/06/2019] [Revised: 11/01/2019] [Accepted: 12/10/2019] [Indexed: 12/11/2022] Open
Abstract
The founding member of the lipoprotein receptor family, low-density lipoprotein receptor (LDLR) plays a major role in the atherogenesis through the receptor-mediated endocytosis of LDL particles and regulation of cholesterol homeostasis. Since the discovery of the LDLR, many other structurally and functionally related receptors have been identified, which include low-density lipoprotein receptor-related protein (LRP)1, LRP5, LRP6, very low-density lipoprotein receptor, and apolipoprotein E receptor 2. The scavenger receptor family members, on the other hand, constitute a family of pattern recognition proteins that are structurally diverse and recognize a wide array of ligands, including oxidized LDL. Among these are cluster of differentiation 36, scavenger receptor class B type I and lectin-like oxidized low-density lipoprotein receptor-1. In addition to the initially assigned role as a mediator of the uptake of macromolecules into the cell, a large number of studies in cultured cells and in in vivo animal models have revealed that these lipoprotein receptors participate in signal transduction to modulate cellular functions. This review highlights the signalling pathways by which these receptors influence the process of atherosclerosis development, focusing on their roles in the vascular cells, such as macrophages, endothelial cells, smooth muscle cells, and platelets. Human genetics of the receptors is also discussed to further provide the relevance to cardiovascular disease risks in humans. Further knowledge of the vascular biology of the lipoprotein receptors and their ligands will potentially enhance our ability to harness the mechanism to develop novel prophylactic and therapeutic strategies against cardiovascular diseases.
Collapse
Affiliation(s)
- Chieko Mineo
- Department of Pediatrics and Cell Biology, Center for Pulmonary and Vascular Biology, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, TX 75390-9063, USA
| |
Collapse
|
12
|
Liu T, Yoon WS, Lee SR. Recent Updates of Lipoprotein(a) and Cardiovascular Disease. Chonnam Med J 2021; 57:36-43. [PMID: 33537217 PMCID: PMC7840349 DOI: 10.4068/cmj.2021.57.1.36] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Revised: 12/14/2020] [Accepted: 12/16/2020] [Indexed: 12/22/2022] Open
Abstract
In recent years, epidemiological studies, genome-wide association studies, and Mendelian randomization studies have shown a strong association between increased levels of lipoproteins and increased risks of coronary heart disease and cardiovascular disease (CVD). Although lipoprotein(a) [Lp(a)] was an independent risk factor for ASCVD, the latest international clinical guidelines do not recommend direct reduction of plasma Lp(a) concentrations. The main reason was that there is no effective clinical medicine that directly lowers plasma Lp(a) concentrations. However, recent clinical trials have shown that proprotein convertase subtilisin/kexin-type 9 inhibitors (PCSK9) and second-generation antisense oligonucleotides can effectively reduce plasma Lp(a) levels. This review will present the structure, pathogenicity, prognostic evidences, and recent advances in therapeutic drugs for Lp(a).
Collapse
Affiliation(s)
- Taili Liu
- Division of Cardiology, Department of Internal Medicine, Chonbuk National University Hospital, Jeonju, Korea
| | - Won-Sik Yoon
- Division of Cardiology, Department of Internal Medicine, Chonbuk National University Hospital, Jeonju, Korea
| | - Sang-Rok Lee
- Division of Cardiology, Department of Internal Medicine, Chonbuk National University Hospital, Jeonju, Korea
| |
Collapse
|
13
|
Cardoso-Saldaña GC, González-Salazar MDC, Posadas-Sánchez R, Vargas-Alarcón G. Metabolic syndrome, lipoprotein(a) and subclinical atherosclerosis in Mexican population. ARCHIVOS DE CARDIOLOGIA DE MEXICO 2021; 91:307-314. [PMID: 33268905 PMCID: PMC8351643 DOI: 10.24875/acm.20000276] [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: 11/30/2022] Open
Abstract
Objective To assess the relationship of metabolic syndrome (MetS) and Lp(a) with subclinical atherosclerosis (CAC) in Mexican adults. Method Clinical, biochemical and tomographic data of visceral, subcutaneous, hepatic abdominal fat and CAC were evaluated in 953 women and men. Lp(a) was determined by nephelometry and MetS was diagnosed according to ATP III criteria. Multivariate logistic regression analysis was performed to determine the independent association of these variables with CAC. Results Age, weight, body mass index, systolic and diastolic blood pressure, volumes of visceral, subcutaneous and hepatic abdominal fat, lipids, glucose, insulin and HOMA-RI were significantly higher in subjects with MetS. The median Lp(a) was lower in subjects with MetS compared to subjects without MetS (3.7 [IR: 2.3-9.2 vs. 5.9 [IR: 2.5-13.1) mg/dL; p < 0.01). The number of components and the MetS were inversely associated with the elevated Lp(a) (> 30 mg / dL). The presence of MetS was associated with a CAC risk >0 (OR: 2.19, [95% CI (1.64-2.94)]; p < 0.001), independently of elevated Lp(a). The components of MetS that were independently associated with the presence of CAC > 0 UA were glycaemia > 100 mg/dL (OR 2.42, [95% CI (1.7-3.4)]; p < 0.0001) and high blood pressure (OR 2.14 [95% CI (1.5-3.1)]; p < 0.0001). Conclusions In Mexican population there is an inverse association between Lp(a) levels and MetS. The MetS and its components were associated with subclinical atherosclerosis. The high prevalence of obesity, diabetes, high blood pressure high triglycerides and low HDL-C, characteristics of Mexican population could explain the differences with other populations.
Collapse
Affiliation(s)
| | | | | | - Gilberto Vargas-Alarcón
- Departamento de Biología Molecular. Instituto Nacional de Cardiología Ignacio Chávez, Ciudad de México, México
| |
Collapse
|
14
|
Huded CP, Shah NP, Puri R, Nicholls SJ, Wolski K, Nissen SE, Cho L. Association of Serum Lipoprotein (a) Levels and Coronary Atheroma Volume by Intravascular Ultrasound. J Am Heart Assoc 2020; 9:e018023. [PMID: 33222598 PMCID: PMC7763761 DOI: 10.1161/jaha.120.018023] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Background Lp(a) (lipoprotein (a)) is a risk factor for cardiovascular events, but the mechanism of increased risk is uncertain. This study evaluated the relationship between Lp(a) and coronary atheroma volume by intravascular ultrasound. Methods and Results This was a post hoc analysis of 6 randomized trials of coronary atheroma by intravascular ultrasound. The population was stratified into high (≥60 mg/dL) and low (<60 mg/dL) baseline serum Lp(a). The primary outcome was baseline coronary percent atheroma volume. A mixed model adjusted for baseline low density lipoprotein, ApoB (apoliporotein B100), non‐high density lipoprotein, sex, age, race, history of myocardial infarction, statin use, and intravascular ultrasound study was used to provide estimates of baseline plaque burden. Of 3943 patients, 17.3% (683) had Lp(a) ≥ 60 mg/dL and 82.7% (3260) had Lp(a) < 60 mg/dL. At baseline, uncorrected low density lipoprotein level (107.7 ± 32.0 versus 99.1 ± 31.5) and statin therapy (99.0% versus 97.0%) were higher in patients with high Lp(a) levels, but low density lipoprotein corrected for Lp(a) was lower (80.6 ± 32.0 versus 94.0 ± 31.4) in patients with high Lp(a) levels. Percent atheroma volume was significantly higher in the high Lp(a) group in unadjusted (38.2% [32.8, 43.6] versus 37.1% [31.4, 43.1], P=0.01) and risk‐adjusted analyses (38.7%±0.5 versus 37.5%±0.5, P<0.001). There was a significant association of increasing risk‐adjusted percent atheroma volume across quintiles of Lp(a) (Lp(a) quintiles 1‐5; 37.3 ± 0.5%, 37.2 ± 0.5%, 37.3 ± 0.5%, 38.0 ± 0.5%, 38.5 ± 0.5%, P=0.002). Conclusions Elevated Lp(a) is independently associated with increased percent atheroma volume. Further work is needed to clarify the relationship of Lp(a)‐lowering treatment with cardiovascular outcomes.
Collapse
|
15
|
Rehberger Likozar A, Zavrtanik M, Šebeštjen M. Lipoprotein(a) in atherosclerosis: from pathophysiology to clinical relevance and treatment options. Ann Med 2020; 52:162-177. [PMID: 32453609 PMCID: PMC7877976 DOI: 10.1080/07853890.2020.1775287] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Lipoprotein(a) (Lp(a)) was discovered more than 50 years ago, and a decade later, it was recognized as a risk factor for coronary artery disease. However, it has gained importance only in the past 10 years, with emergence of drugs that can effectively decrease its levels. Lp(a) is a low-density lipoprotein (LDL) with an added apolipoprotein(a) attached to the apolipoprotein B component via a disulphide bond. Circulating levels of Lp(a) are mainly genetically determined. Lp(a) has many functions, which include proatherosclerotic, prothrombotic and pro-inflammatory roles. Here, we review recent data on the role of Lp(a) in the atherosclerotic process, and treatment options for patients with cardiovascular diseases. Currently 'Proprotein convertase subtilisin/kexin type 9' (PCSK9) inhibitors that act through non-specific reduction of Lp(a) are the only drugs that have shown effectiveness in clinical trials, to provide reductions in cardiovascular morbidity and mortality. The effects of PCSK9 inhibitors are not purely through Lp(a) reduction, but also through LDL cholesterol reduction. Finally, we discuss new drugs on the horizon, and gene-based therapies that affect transcription and translation of apolipoprotein(a) mRNA. Clinical trials in patients with high Lp(a) and low LDL cholesterol might tell us whether Lp(a) lowering per se decreases cardiovascular morbidity and mortality.KEY MESSAGESLipoprotein(a) is an important risk factor in patients with cardiovascular diseases.Lipoprotein(a) has many functions, which include proatherosclerotic, prothrombotic and pro-inflammatory roles.Treatment options to lower lipoprotein(a) levels are currently scarce, but new drugs are on the horizon.
Collapse
Affiliation(s)
| | - Mark Zavrtanik
- Division of Internal Medicine, University Medical Centre Ljubljana, Ljubljana, Slovenia
| | - Miran Šebeštjen
- Department of Vascular Diseases, University Medical Centre Ljubljana, Ljubljana, Slovenia.,Department of Cardiology, University Medical Centre Ljubljana, Ljubljana, Slovenia.,Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| |
Collapse
|
16
|
Familial Hypercholesterolemia and Lipoprotein(a). J Am Coll Cardiol 2020; 75:2694-2697. [DOI: 10.1016/j.jacc.2020.04.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Accepted: 04/01/2020] [Indexed: 12/24/2022]
|
17
|
Zhang X, Stiekema LCA, Stroes ESG, Groen AK. Metabolic effects of PCSK9 inhibition with Evolocumab in subjects with elevated Lp(a). Lipids Health Dis 2020; 19:91. [PMID: 32393252 PMCID: PMC7216641 DOI: 10.1186/s12944-020-01280-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Accepted: 05/06/2020] [Indexed: 11/16/2022] Open
Abstract
Background Epidemiological studies substantiated that subjects with elevated lipoprotein(a) [Lp(a)] have a markedly increased cardiovascular risk. Inhibition of proprotein convertase subtilisin/kexin type 9 (PCSK9) lowers both LDL cholesterol (LDL-C) as well as Lp(a), albeit modestly. Effects of PCSK9 inhibition on circulating metabolites such as lipoprotein subclasses, amino acids and fatty acids remain to be characterized. Methods We performed nuclear magnetic resonance (NMR) metabolomics on plasma samples derived from 30 individuals with elevated Lp(a) (> 150 mg/dL). The 30 participants were randomly assigned into two groups, placebo (N = 14) and evolocumab (N = 16). We assessed the effect of 16 weeks of evolocumab 420 mg Q4W treatment on circulating metabolites by running lognormal regression analyses, and compared this to placebo. Subsequently, we assessed the interrelationship between Lp(a) and 14 lipoprotein subclasses in response to treatment with evolocumab, by running multilevel multivariate regression analyses. Results On average, evolocumab treatment for 16 weeks resulted in a 17% (95% credible interval: 8 to 26%, P < 0.001) reduction of circulating Lp(a), coupled with substantial reduction of VLDL, IDL and LDL particles as well as their lipid contents. Interestingly, increasing concentrations of baseline Lp(a) were associated with larger reduction in triglyceride-rich VLDL particles after evolocumab treatment. Conclusions Inhibition of PCSK9 with evolocumab markedly reduced VLDL particle concentrations in addition to lowering LDL-C. The extent of reduction in VLDL particles depended on the baseline level of Lp(a). Our findings suggest a marked effect of evolocumab on VLDL metabolism in subjects with elevated Lp(a). Trial registration Clinical trial registration information is registered at ClinicalTrials.gov on April 14, 2016 with the registration number NCT02729025.
Collapse
Affiliation(s)
- Xiang Zhang
- Department of Experimental Vascular Medicine, Amsterdam University Medical Center, University of Amsterdam, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands. .,Human and Animal Physiology, Wageningen University, De Elst 1, 6708 WD, Wageningen, The Netherlands.
| | - Lotte C A Stiekema
- Department of Vascular Medicine, Amsterdam University Medical Center, University of Amsterdam, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands
| | - Erik S G Stroes
- Department of Vascular Medicine, Amsterdam University Medical Center, University of Amsterdam, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands
| | - Albert K Groen
- Department of Experimental Vascular Medicine, Amsterdam University Medical Center, University of Amsterdam, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands
| |
Collapse
|
18
|
zahedi avval F, Moammeri Salahshooh M, sadabadi F. Lipoprotein(a) and its Clinical Importance. MEDICAL LABORATORY JOURNAL 2020. [DOI: 10.29252/mlj.14.2.1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022] Open
|
19
|
Boffa MB, Koschinsky ML. Proprotein convertase subtilisin/kexin type 9 inhibitors and lipoprotein(a)-mediated risk of atherosclerotic cardiovascular disease: more than meets the eye? Curr Opin Lipidol 2019; 30:428-437. [PMID: 31577611 DOI: 10.1097/mol.0000000000000641] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
PURPOSE OF REVIEW Evidence continues to mount for elevated lipoprotein(a) [Lp(a)] as a prevalent, independent, and causal risk factor for atherosclerotic cardiovascular disease. However, the effects of existing lipid-lowering therapies on Lp(a) are comparatively modest and are not specific to Lp(a). Consequently, evidence that Lp(a)-lowering confers a cardiovascular benefit is lacking. Large-scale cardiovascular outcome trials (CVOTs) of inhibitory mAbs targeting proprotein convertase subtilisin/kexin type 9 inhibitors (PCSK9i) may address this issue. RECENT FINDINGS Although the ability of PCSK9i to lower Lp(a) by 15-30% is now clear, the mechanisms involved continue to be debated, with in-vitro and in-vivo studies showing effects on Lp(a) clearance (through the LDL receptor or other receptors) and Lp(a)/apolipoprotein(a) biosynthesis in hepatocytes. The FOURIER CVOT showed that patients with higher baseline levels of Lp(a) derived greater benefit from evolocumab and those with the lowest combined achieved Lp(a) and LDL-cholesterol (LDL-C) had the lowest event rate. Meta-analysis of ten phase 3 trials of alirocumab came to qualitatively similar conclusions concerning achieved Lp(a) levels, although an effect independent of LDL-C lowering could not be demonstrated. SUMMARY Although it is not possible to conclude that PCSK9i specifically lower Lp(a)-attributable risk, patients with elevated Lp(a) could derive incremental benefit from PCSK9i therapy.
Collapse
Affiliation(s)
| | - Marlys L Koschinsky
- Department of Physiology & Pharmacology
- Robarts Research Institute, Schulich School of Medicine & Dentistry, The University of Western Ontario, London, Ontario, Canada
| |
Collapse
|
20
|
Mihăilă RG. Pragmatic Analysis of Dyslipidemia Involvement in Coronary Artery Disease: A Narrative Review. Curr Cardiol Rev 2019; 16:36-47. [PMID: 31113345 PMCID: PMC7393591 DOI: 10.2174/1573403x15666190522100041] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/03/2019] [Revised: 05/06/2019] [Accepted: 05/07/2019] [Indexed: 11/22/2022] Open
Abstract
Background Dyslipidemia is the main factor involved in the occurrence and progression of coronary artery disease. Objective The research strategy is aimed at analyzing new data on the pathophysiology of dyslipidemia involvement in coronary artery disease, the modalities of atherogenic risk estimation and therapeutic advances. Methods Scientific articles published in PubMed from January 2017 to February 2018 were searched using the terms “dyslipidemia” and “ischemic heart disease”. Results PCSK9 contributes to the increase in serum levels of low-density lipoprotein-cholesterol and lipoprotein (a). The inflammation is involved in the progression of hyperlipidemia and atherosclerosis. Hypercholesterolemia changes the global cardiac gene expression profile and is thus involved in the increase of oxidative stress, mitochondrial dysfunction, and apoptosis initiated by inflammation. Coronary artery calcifications may estimate the risk of coronary events. The cardio-ankle vascular index evaluates the arterial stiffness and correlates with subclinical coronary atherosclerosis. The carotid plaque score is superior to carotid intima-media thickness for risk stratification in patients with familial hypercholesterolemia and both can independently predict coronary artery disease. The lipoprotein (a) and familial hypercholesterolemia have a synergistic role in predicting the risk of early onset and severity of coronary atherosclerosis. A decrease in atherosclerotic coronary plaque progression can be achieved in patients with plasma LDL-cholesterol levels below 70 mg/dL. A highly durable RNA interference therapeutic inhibitor of PCSK9 synthesis could be a future solution. Conclusion The prophylaxis and treatment of coronary artery disease in a dyslipidemic patient should be based on a careful assessment of cardio-vascular risk factors and individual metabolic particularities, so it may be personalized.
Collapse
Affiliation(s)
- Romeo-Gabriel Mihăilă
- Faculty of Medicine, "Lucian Blaga" University of Sibiu, Sibiu, Romania; CVASIC Laboratory, Emergency County Clinical Hospital Sibiu, Sibiu, Romania
| |
Collapse
|
21
|
Nakajima K, Tokita Y, Tanaka A, Takahashi S. The VLDL receptor plays a key role in the metabolism of postprandial remnant lipoproteins. Clin Chim Acta 2019; 495:382-393. [PMID: 31078566 DOI: 10.1016/j.cca.2019.05.004] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2019] [Revised: 05/03/2019] [Accepted: 05/06/2019] [Indexed: 12/21/2022]
Abstract
A new concept to account for the process of postprandial remnant lipoprotein metabolism is proposed based on the characteristics of lipoprotein particles and their receptors. The characteristics of remnant lipoprotein (RLP) were investigated using an immuno-separation method. The majority of the postprandial lipoproteins increased after fat intake was shown to be VLDL remnants, not chylomicron (CM) remnants, based on the significantly high ratio of apoB100/apoB48 in the RLP and the high degree of similarity in the particle size of the apoB48 and apoB100 carrying lipoproteins, which fluctuate in parallel during a 6 h period after fat intake. The VLDL receptor was discovered as a receptor for TG-rich lipoprotein metabolism and is located in peripheral tissues such as skeletal muscle, adipose tissue, etc., but not in the liver. Postprandial VLDL particles are strongly bound and internalized into cells expressing the VLDL receptor. Ligands that bind to VLDL receptor, such as LPL and Lp(a), present in RLP. The presence of various specific ligands in VLDL remnants may enhance the capacity for binding to the VLDL receptor, which play the role primarily for energy delivery to the peripheral tissues, but is also a causal factor in atherogenic diseases when excessively and/or continuously remained in plasma.
Collapse
Affiliation(s)
- Katsuyuki Nakajima
- Laboratory of Clinical Nutrition and Medicine, Kagawa Nutrition University, Tokyo, Japan; Graduate School of Health Sciences, Gunma University, Maebashi, Gunma, Japan.
| | - Yoshiharu Tokita
- Laboratory of Clinical Nutrition and Medicine, Kagawa Nutrition University, Tokyo, Japan; Graduate School of Health Sciences, Gunma University, Maebashi, Gunma, Japan
| | - Akira Tanaka
- Laboratory of Clinical Nutrition and Medicine, Kagawa Nutrition University, Tokyo, Japan
| | - Sadao Takahashi
- Laboratory of Clinical Nutrition and Medicine, Kagawa Nutrition University, Tokyo, Japan; Division of Diabetes, Ageo Central General Hospital, Saitama, Japan
| |
Collapse
|
22
|
Borrelli MJ, Youssef A, Boffa MB, Koschinsky ML. New Frontiers in Lp(a)-Targeted Therapies. Trends Pharmacol Sci 2019; 40:212-225. [PMID: 30732864 DOI: 10.1016/j.tips.2019.01.004] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2018] [Revised: 01/07/2019] [Accepted: 01/08/2019] [Indexed: 12/13/2022]
Abstract
Interest in lipoprotein (a) [Lp(a)] has exploded over the past decade with the emergence of genetic and epidemiological studies pinpointing elevated levels of this unique lipoprotein as a causal risk factor for atherosclerotic cardiovascular disease (ASCVD) and calcific aortic valve disease (CAVD). This review summarizes the most recent discoveries regarding therapeutic approaches to lower Lp(a) and presents these findings in the context of an emerging, although far from complete, understanding of the biosynthesis and catabolism of Lp(a). Application of Lp(a)-specific lowering agents to outcome trials will be the key to opening this new frontier in the battle against CVD.
Collapse
Affiliation(s)
- Matthew J Borrelli
- Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, The University of Western Ontario, London, Ontario, Canada
| | - Amer Youssef
- Robarts Research Institute, Schulich School of Medicine and Dentistry, The University of Western Ontario, London, Ontario, Canada
| | - Michael B Boffa
- Robarts Research Institute, Schulich School of Medicine and Dentistry, The University of Western Ontario, London, Ontario, Canada; Department of Biochemistry, Schulich School of Medicine and Dentistry, The University of Western Ontario, London, Ontario, Canada
| | - Marlys L Koschinsky
- Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, The University of Western Ontario, London, Ontario, Canada; Robarts Research Institute, Schulich School of Medicine and Dentistry, The University of Western Ontario, London, Ontario, Canada.
| |
Collapse
|
23
|
Ma L, Waldmann E, Ooi EMM, Chan DC, Barrett HPR, Watts GF, Parhofer KG. Lipoprotein (a) and Low-density lipoprotein apolipoprotein B metabolism following apheresis in patients with elevated lipoprotein(a) and coronary artery disease. Eur J Clin Invest 2019; 49:e13053. [PMID: 30447089 DOI: 10.1111/eci.13053] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/24/2018] [Revised: 11/04/2018] [Accepted: 11/13/2018] [Indexed: 11/30/2022]
Abstract
BACKGROUND Lipoprotein apheresis effectively lowers lipoprotein(a) [Lp(a)] and low-density lipoprotein (LDL) by approximately 60%-70%. The rebound of LDL and Lp(a) particle concentrations following lipoprotein apheresis allows the determination of fractional catabolic rate (FCR) and hence production rate (PR) during non-steady state conditions. We aimed to investigate the kinetics of Lp(a) and LDL apolipoprotein B-100 (apoB) particles in patients with elevated Lp(a) and coronary artery disease undergoing regular apheresis. PATIENTS AND METHODS A cross-sectional study was carried out in 13 patients with elevated Lp(a) concentration (>500 mg/L) and coronary artery disease. Lp(a) and LDL-apoB metabolic parameters, including FCR and PR were derived by the fit of a compartment model to the Lp(a) and LDL-apoB concentration data following lipoprotein apheresis. RESULTS The FCR of Lp(a) was significantly lower than that of LDL-apoB (0.39 [0.31, 0.49] vs 0.57 [0.46, 0.71] pools/day, P = 0.03) with no significant differences in the corresponding PR (14.80 [11.34, 19.32] vs 15.73 [11.93, 20.75] mg/kg/day, P = 0.80). No significant associations were observed between the FCR and PR of Lp(a) and LDL-apoB. CONCLUSIONS In patients with elevated Lp(a), the fractional catabolism of Lp(a) is slower than that of LDL-apoB particles, implying that different metabolic pathways are involved in the catabolism of these lipoproteins. These findings have implications for new therapies for lowering apolipoprotein(a) and apoB to prevent atherosclerotic cardiovascular disease.
Collapse
Affiliation(s)
- Louis Ma
- School of Biomedical Science, University of Western Australia, Western Australia, Australia.,School of Medicine, University of Western Australia, Western Australia, Australia
| | - Elisa Waldmann
- Department of Internal Medicine IV, Ludwig-Maximilans-University Munich, Munich, Germany
| | - Esther M M Ooi
- School of Biomedical Science, University of Western Australia, Western Australia, Australia
| | - Dick C Chan
- School of Biomedical Science, University of Western Australia, Western Australia, Australia.,School of Medicine, University of Western Australia, Western Australia, Australia
| | - Hugh P R Barrett
- School of Biomedical Science, University of Western Australia, Western Australia, Australia
| | - Gerald F Watts
- School of Medicine, University of Western Australia, Western Australia, Australia.,Department of Cardiology, Lipid Disorders Clinic, Royal Perth Hospital, Western Australia, Australia
| | - Klaus G Parhofer
- Department of Internal Medicine IV, Ludwig-Maximilans-University Munich, Munich, Germany
| |
Collapse
|
24
|
McCormick SPA, Schneider WJ. Lipoprotein(a) catabolism: a case of multiple receptors. Pathology 2018; 51:155-164. [PMID: 30595508 DOI: 10.1016/j.pathol.2018.11.003] [Citation(s) in RCA: 66] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2018] [Revised: 10/31/2018] [Accepted: 11/01/2018] [Indexed: 01/09/2023]
Abstract
Lipoprotein(a) [Lp(a)] is an apolipoprotein B (apoB)-containing plasma lipoprotein similar in structure to low-density lipoprotein (LDL). Lp(a) is more complex than LDL due to the presence of apolipoprotein(a) [apo(a)], a large glycoprotein sharing extensive homology with plasminogen, which confers some unique properties onto Lp(a) particles. ApoB and apo(a) are essential for the assembly and catabolism of Lp(a); however, other proteins associated with the particle may modify its metabolism. Lp(a) specifically carries a cargo of oxidised phospholipids (OxPL) bound to apo(a) which stimulates many proinflammatory pathways in cells of the arterial wall, a key property underlying its pathogenicity and association with cardiovascular disease (CVD). While the liver and kidney are the major tissues implicated in Lp(a) clearance, the pathways for Lp(a) uptake appear to be complex and are still under investigation. Biochemical studies have revealed an exceptional array of receptors that associate with Lp(a) either via its apoB, apo(a), or OxPL components. These receptors fall into five main categories, namely 'classical' lipoprotein receptors, toll-like and scavenger receptors, lectins, and plasminogen receptors. The roles of these receptors have largely been dissected by genetic manipulation in cells or mice, although their relative physiological importance for removal of Lp(a) from the circulation remains unclear. The LPA gene encoding apo(a) has an overwhelming effect on Lp(a) levels which precludes any clear associations between potential Lp(a) receptor genes and Lp(a) levels in population studies. Targeted approaches and the selection of unique Lp(a) phenotypes within populations has nevertheless allowed for some associations to be made. Few of the proposed Lp(a) receptors can specifically be manipulated with current drugs and, as such, it is not currently clear whether any of these receptors could provide relevant targets for therapeutic manipulation of Lp(a) levels. This review summarises the current status of knowledge about receptor-mediated pathways for Lp(a) catabolism.
Collapse
Affiliation(s)
- Sally P A McCormick
- Department of Biochemistry, School of Biomedical Sciences, University of Otago, Dunedin, New Zealand.
| | - Wolfgang J Schneider
- Department of Medical Biochemistry, Max F. Perutz Laboratories, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
25
|
Najjar RS, Moore CE, Montgomery BD. Consumption of a defined, plant-based diet reduces lipoprotein(a), inflammation, and other atherogenic lipoproteins and particles within 4 weeks. Clin Cardiol 2018; 41:1062-1068. [PMID: 30014498 DOI: 10.1002/clc.23027] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2018] [Revised: 07/07/2018] [Accepted: 07/12/2018] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND Lipoprotein(a) [Lp(a)] is a highly atherogenic lipoprotein and is minimally effected by lifestyle changes. While some drugs can reduce Lp(a), diet has not consistently shown definitive reduction of this biomarker. The effect of consuming a plant-based diet on serum Lp(a) concentrations have not been previously evaluated. HYPOTHESIS Consumption of a defined, plant-based for 4 weeks reduces Lp(a). METHODS Secondary analysis of a previous trial was conducted, in which overweight and obese individuals (n = 31) with low-density lipoprotein cholesterol concentrations >100 mg/dL consumed a defined, plant-based diet for 4 weeks. Baseline and 4-week labs were collected. Data were analyzed using a paired samples t-test. RESULTS Significant reductions were observed for serum Lp(a) (-32.0 ± 52.3 nmol/L, P = 0.003), apolipoprotein B (-13.2 ± 18.3 mg/dL, P < 0.0005), low-density lipoprotein (LDL) particles (-304.8 ± 363.0 nmol/L, P < 0.0005) and small-dense LDL cholesterol (-10.0 ± 9.2 mg/dL, P < 0.0005). Additionally, serum interleukin-6 (IL-6), total white blood cells, lipoprotein-associated phospholipase A2 (Lp-PLA2), high-sensitivity c-reactive protein (hs-CRP), and fibrinogen were significantly reduced (P ≤ 0.004). CONCLUSIONS A defined, plant-based diet has a favorable impact on Lp(a), inflammatory indicators, and other atherogenic lipoproteins and particles. Lp(a) concentration was previously thought to be only minimally altered by dietary interventions. In this protocol however, a defined plant-based diet was shown to substantially reduce this biomarker. Further investigation is required to elucidate the specific mechanisms that contribute to the reductions in Lp(a) concentrations, which may include alterations in gene expression.
Collapse
Affiliation(s)
- Rami S Najjar
- Department of Nutrition, Georgia State University, Atlanta, Georgia
| | - Carolyn E Moore
- Department of Nutrition and Food Science, Texas Woman's University, Houston, Texas
| | - Baxter D Montgomery
- University of Texas Health Science Center, Houston, Texas.,Montgomery Heart & Wellness, Houston, Texas
| |
Collapse
|
26
|
Kostner KM, Kostner GM, Wierzbicki AS. Is Lp(a) ready for prime time use in the clinic? A pros-and-cons debate. Atherosclerosis 2018; 274:16-22. [PMID: 29747086 DOI: 10.1016/j.atherosclerosis.2018.04.032] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/12/2018] [Revised: 04/16/2018] [Accepted: 04/25/2018] [Indexed: 12/11/2022]
Abstract
Lipoprotein (a) (Lp(a)) is a cholesterol-rich lipoprotein known since 1963. In spite of extensive research on Lp(a), there are still numerous gaps in our knowledge relating to its function, biosynthesis and catabolism. One reason for this might be that apo(a), the characteristic glycoprotein of Lp(a), is expressed only in primates. Results from experiments using transgenic animals therefore may need verification in humans. Studies on Lp(a) are also handicapped by the great number of isoforms of apo(a) and the heterogeneity of apo(a)-containing fractions in plasma. Quantification of Lp(a) in the clinical laboratory for a long time has not been standardized. Starting from its discovery, reports accumulated that Lp(a) contributed to the risk of cardiovascular disease (CVD), myocardial infarction (MI) and stroke. Early reports were based on case control studies but in the last decades a great deal of prospective studies have been published that highlight the increased risk for CVD and MI in patients with elevated Lp(a). Final answers to the question of whether Lp(a) is ready for wider clinical use will come from intervention studies with novel selective Lp(a) lowering medications that are currently underway. This article expounds arguments for and against this proposition from currently available data.
Collapse
Affiliation(s)
- Karam M Kostner
- Department of Cardiology, Mater Hospital and University of Queensland, Brisbane, Australia
| | - Gert M Kostner
- Department of Molecular Biology and Biochemistry, Gottfried Schatz Research Center for Cell Signaling, Medical University of Graz, Austria
| | - Anthony S Wierzbicki
- Department of Metabolic Medicine/Chemical Pathology, Guy's & St Thomas' Hospitals, London, UK.
| |
Collapse
|
27
|
Zanoni P, Velagapudi S, Yalcinkaya M, Rohrer L, von Eckardstein A. Endocytosis of lipoproteins. Atherosclerosis 2018; 275:273-295. [PMID: 29980055 DOI: 10.1016/j.atherosclerosis.2018.06.881] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/16/2018] [Revised: 06/04/2018] [Accepted: 06/22/2018] [Indexed: 02/06/2023]
Abstract
During their metabolism, all lipoproteins undergo endocytosis, either to be degraded intracellularly, for example in hepatocytes or macrophages, or to be re-secreted, for example in the course of transcytosis by endothelial cells. Moreover, there are several examples of internalized lipoproteins sequestered intracellularly, possibly to exert intracellular functions, for example the cytolysis of trypanosoma. Endocytosis and the subsequent intracellular itinerary of lipoproteins hence are key areas for understanding the regulation of plasma lipid levels as well as the biological functions of lipoproteins. Indeed, the identification of the low-density lipoprotein (LDL)-receptor and the unraveling of its transcriptional regulation led to the elucidation of familial hypercholesterolemia as well as to the development of statins, the most successful therapeutics for lowering of cholesterol levels and risk of atherosclerotic cardiovascular diseases. Novel limiting factors of intracellular trafficking of LDL and the LDL receptor continue to be discovered and to provide drug targets such as PCSK9. Surprisingly, the receptors mediating endocytosis of high-density lipoproteins or lipoprotein(a) are still a matter of controversy or even new discovery. Finally, the receptors and mechanisms, which mediate the uptake of lipoproteins into non-degrading intracellular itineraries for re-secretion (transcytosis, retroendocytosis), storage, or execution of intracellular functions, are largely unknown.
Collapse
Affiliation(s)
- Paolo Zanoni
- Institute for Clinical Chemistry, University and University Hospital Zurich, Zurich, Switzerland; Centre for Integrative Human Physiology, University of Zurich, Zurich, Switzerland
| | - Srividya Velagapudi
- Institute for Clinical Chemistry, University and University Hospital Zurich, Zurich, Switzerland; Centre for Integrative Human Physiology, University of Zurich, Zurich, Switzerland
| | - Mustafa Yalcinkaya
- Institute for Clinical Chemistry, University and University Hospital Zurich, Zurich, Switzerland; Centre for Integrative Human Physiology, University of Zurich, Zurich, Switzerland
| | - Lucia Rohrer
- Institute for Clinical Chemistry, University and University Hospital Zurich, Zurich, Switzerland; Centre for Integrative Human Physiology, University of Zurich, Zurich, Switzerland
| | - Arnold von Eckardstein
- Institute for Clinical Chemistry, University and University Hospital Zurich, Zurich, Switzerland; Centre for Integrative Human Physiology, University of Zurich, Zurich, Switzerland.
| |
Collapse
|
28
|
Watts GF, Chan DC, Somaratne R, Wasserman SM, Scott R, Marcovina SM, Barrett PHR. Controlled study of the effect of proprotein convertase subtilisin-kexin type 9 inhibition with evolocumab on lipoprotein(a) particle kinetics. Eur Heart J 2018; 39:2577-2585. [DOI: 10.1093/eurheartj/ehy122] [Citation(s) in RCA: 91] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/10/2017] [Accepted: 03/02/2018] [Indexed: 12/24/2022] Open
Affiliation(s)
- Gerald F Watts
- Lipid Disorders Clinic, Department of Cardiology, Royal Perth Hospital, Perth, WA, Australia
- Schools of Medicine and Biomedical Science, University of Western Australia, Perth, WA, Australia
| | - Dick C Chan
- Schools of Medicine and Biomedical Science, University of Western Australia, Perth, WA, Australia
| | | | | | - Rob Scott
- Formerly of Amgen, Inc., Thousand Oaks, CA, USA
| | - Santica M Marcovina
- Northwest Lipid Metabolism and Diabetes Research Laboratories, Division of Metabolism, Endocrinology, and Nutrition, Department of Medicine, University of Washington, Seattle, WA, USA
| | - P Hugh R Barrett
- Schools of Medicine and Biomedical Science, University of Western Australia, Perth, WA, Australia
| |
Collapse
|
29
|
Page MM, Watts GF. PCSK9 in context: A contemporary review of an important biological target for the prevention and treatment of atherosclerotic cardiovascular disease. Diabetes Obes Metab 2018; 20:270-282. [PMID: 28736830 DOI: 10.1111/dom.13070] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2017] [Revised: 07/19/2017] [Accepted: 07/19/2017] [Indexed: 12/16/2022]
Abstract
The identification of the critical role of proprotein convertase subtilisin/kexin type 9 (PCSK9) has rapidly led to the development of PCSK9 inhibition with monoclonal antibodies (mAbs). PCSK9 mAbs are already in limited clinical use and are the subject of major cardiovascular outcomes trials, which, if universally positive, could see much wider clinical application of these agents. Patients with familial hypercholesterolaemia are the most obvious candidates for these drugs, but other patients with elevated cardiovascular risk, statin intolerance or hyperlipoproteinaemia(a) may also benefit. PCSK9 mAbs, administered once or twice monthly, reduce LDL cholesterol levels by 50% to 70%, and appear to be safe and acceptable to patients over at least 2 years of treatment; however, treatment-emergent adverse effects are not always identified in clinical trials, as well-evidenced by statin myopathy. Inclisiran is a promising RNA-based therapy that promotes the degradation of PCSK9 mRNA transcripts and has similar efficacy to mAbs, but with a much longer duration of action. The cost-effectiveness and long-term safety of therapies targeted at inhibiting PCSK9 remain to be demonstrated if they are to be used widely in coronary prevention.
Collapse
Affiliation(s)
- Michael M Page
- Department of Clinical Biochemistry, PathWest Laboratory Medicine, Fiona Stanley Hospital, Perth, Western Australia
- School of Medicine, Faculty of Health and Medical Sciences, University of Western Australia, Perth, Western Australia
| | - Gerald F Watts
- Lipid Disorders Clinic, Department of Cardiology, Royal Perth Hospital, Perth, Western Australia
- School of Medicine, Faculty of Health and Medical Sciences, University of Western Australia, Perth, Western Australia
| |
Collapse
|
30
|
Nakajima K, Tanaka A. Atherogenic postprandial remnant lipoproteins; VLDL remnants as a causal factor in atherosclerosis. Clin Chim Acta 2018; 478:200-215. [PMID: 29307667 DOI: 10.1016/j.cca.2017.12.039] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2017] [Revised: 12/23/2017] [Accepted: 12/24/2017] [Indexed: 01/02/2023]
Abstract
Oxidized LDL (Ox-LDL) and chylomicron (CM) remnants have been suggested to be the most atherogenic lipoproteins that initiate and exacerbate coronary atherosclerosis. In this review, we propose a hypothesis of the causal lipoproteins in atherosclerosis based on our recent findings on postprandial remnant lipoproteins (RLP). Plasma RLP-C and RLP-TG increased significantly after food intake, especially a fat load. More than 80% of the TG increase after the fat load consisted of the TG in RLP, which contained significantly greater apoB100 than apoB48 particles as VLDL remnants. The majority of the LPL in non-heparin plasma was found in RLP as an RLP-LPL complex and released into the circulation after hydrolysis. Plasma LPL did not increase after food intake, which may have caused the partial hydrolysis of CM and VLDL as well as the significant increase of RLP-TG in the postprandial plasma. LPL was inversely correlated with the RLP particle size after food intake. We showed that VLDL remnants are the major atherogenic lipoproteins in the postprandial plasma associated with insufficient LPL activity and a causal factor in the initiation and progression of atherosclerosis. We also propose "LPL bound TG-rich lipoproteins" as a new definition of remnant lipoproteins based on the findings of the RLP-LPL complex in the non-heparin plasma.
Collapse
Affiliation(s)
- Katsuyuki Nakajima
- Laboratory of Clinical Nutrition and Medicine, Kagawa Nutrition University, Tokyo, Japan; Department of Clinical Laboratory Medicine, Gunma University Graduate School of Medicine, Maebashi, Japan.
| | - Akira Tanaka
- Laboratory of Clinical Nutrition and Medicine, Kagawa Nutrition University, Tokyo, Japan
| |
Collapse
|
31
|
Scipione CA, Koschinsky ML, Boffa MB. Lipoprotein(a) in clinical practice: New perspectives from basic and translational science. Crit Rev Clin Lab Sci 2017; 55:33-54. [PMID: 29262744 DOI: 10.1080/10408363.2017.1415866] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Elevated plasma concentrations of lipoprotein(a) (Lp(a)) are a causal risk factor for coronary heart disease (CHD) and calcific aortic valve stenosis (CAVS). Genetic, epidemiological and in vitro data provide strong evidence for a pathogenic role for Lp(a) in the progression of atherothrombotic disease. Despite these advancements and a race to develop new Lp(a) lowering therapies, there are still many unanswered and emerging questions about the metabolism and pathophysiology of Lp(a). New studies have drawn attention to Lp(a) as a contributor to novel pathogenic processes, yet the mechanisms underlying the contribution of Lp(a) to CVD remain enigmatic. New therapeutics show promise in lowering plasma Lp(a) levels, although the complete mechanisms of Lp(a) lowering are not fully understood. Specific agents targeted to apolipoprotein(a) (apo(a)), namely antisense oligonucleotide therapy, demonstrate potential to decrease Lp(a) to levels below the 30-50 mg/dL (75-150 nmol/L) CVD risk threshold. This therapeutic approach should aid in assessing the benefit of lowering Lp(a) in a clinical setting.
Collapse
Affiliation(s)
- Corey A Scipione
- a Department of Advanced Diagnostics , Toronto General Hospital Research Institute, UHN , Toronto , Canada
| | - Marlys L Koschinsky
- b Robarts Research Institute , Western University , London , Canada.,c Department of Physiology & Pharmacology , Schulich School of Medicine & Dentistry, Western University , London , Canada
| | - Michael B Boffa
- d Department of Biochemistry , Western University , London , Canada
| |
Collapse
|
32
|
Abstract
PURPOSE OF REVIEW Genetic dyslipidemias contribute to the prevalence of ischemic heart disease. The field of genetic dyslipidemias and their influence on atherosclerotic heart disease is rapidly developing and accumulating increasing evidence. The purpose of this review is to describe the current state of knowledge in regard to inherited atherogenic dyslipidemias. The disorders of familial hypercholesterolemia (FH) and elevated lipoprotein(a) will be detailed. Genetic technology has made rapid advancements, leading to new discoveries in inherited atherogenic dyslipidemias, which will be explored in this review, as well as a description of possible future developments. Increasing attention has come upon the genetic disorders of familial hypercholesterolemia and elevated lipoprotein(a). RECENT FINDINGS This review includes new knowledge of these disorders including description of these disorders, their method of diagnosis, their prevalence, their genetic underpinnings, and their effect on the development of cardiovascular disease. In addition, it discusses major advances in genetic technology, including the completion of the human genome sequence, next-generation sequencing, and genome-wide association studies. Also discussed are rare variant studies with specific genetic mechanisms involved in inherited dyslipidemias, such as in the proprotein convertase subtilisin/kexin type 9 (PCSK9) enzyme. The field of genetics of dyslipidemia and cardiovascular disease is rapidly growing, which will result in a bright future of novel mechanisms of action and new therapeutics.
Collapse
Affiliation(s)
- Kavita Sharma
- Ohio Health Heart and Vascular Physicians, 765 North Hamilton Road, Suite 120, Gahanna, OH, 43230, USA
| | - Ragavendra R Baliga
- The Ohio State University Wexner Medical Center, Suite 200, 473 West 12th Avenue, Columbus, OH, 43210, USA.
| |
Collapse
|
33
|
Ellis KL, Boffa MB, Sahebkar A, Koschinsky ML, Watts GF. The renaissance of lipoprotein(a): Brave new world for preventive cardiology? Prog Lipid Res 2017; 68:57-82. [DOI: 10.1016/j.plipres.2017.09.001] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2017] [Revised: 09/01/2017] [Accepted: 09/05/2017] [Indexed: 12/24/2022]
|
34
|
Hishikari K, Hikita H, Nakamura S, Nakagama S, Mizusawa M, Yamamoto T, Doi J, Utsugi Y, Sudo Y, Kimura S, Ashikaga T, Takahashi A, Isobe M. Usefulness of Lipoprotein(a) for Predicting Clinical Outcomes After Endovascular Therapy for Aortoiliac Atherosclerotic Lesions. J Endovasc Ther 2017; 24:793-799. [PMID: 28830274 DOI: 10.1177/1526602817728068] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
PURPOSE To evaluate the usefulness of serum lipoprotein(a) as a biomarker of clinical outcomes after endovascular therapy (EVT) for atherosclerotic aortoiliac lesions. METHODS Serum lipoprotein(a) concentrations were measured at admission in 189 consecutive patients (median age 72 years; 160 men) with peripheral artery disease who underwent EVT for aortoiliac occlusive disease. The patients were dichotomized into 2 groups based on serum lipoprotein(a) levels ≤40 mg/dL (LOW; n=135) or >40 mg/dL (HIGH; n=54). After EVT, the incidences of major adverse limb events (MALE) were analyzed. Predictors of MALE were sought with a Cox proportional hazards analysis; results are presented as the hazard ratio (HR) and 95% confidence interval. RESULTS At the median follow-up of 33 months (interquartile range 11, 54), MALE occurred in 44 (23.3%) patients. The MALE-free survival estimate was significantly lower in patients in the HIGH group (55.6% vs 85.2%, p<0.001). Independent predictors of MALE after EVT were hemodialysis (HR 2.23, 95% CI 1.04 to 4.78, p=0.039) and high lipoprotein(a) levels (HR 2.80, 95% CI 1.44 to 5.45, p=0.003). CONCLUSION High lipoprotein(a) levels were associated with a higher incidence of MALE after EVT for patients with aortoiliac lesions.
Collapse
Affiliation(s)
- Keiichi Hishikari
- 1 Cardiovascular Center, Yokosuka Kyosai Hospital, Yokosuka, Japan.,2 Cardiovascular Medicine, Tokyo Medical and Dental University, Tokyo, Japan
| | - Hiroyuki Hikita
- 1 Cardiovascular Center, Yokosuka Kyosai Hospital, Yokosuka, Japan
| | - Shun Nakamura
- 1 Cardiovascular Center, Yokosuka Kyosai Hospital, Yokosuka, Japan
| | - Shun Nakagama
- 1 Cardiovascular Center, Yokosuka Kyosai Hospital, Yokosuka, Japan
| | | | - Tasuku Yamamoto
- 1 Cardiovascular Center, Yokosuka Kyosai Hospital, Yokosuka, Japan
| | - Junichi Doi
- 1 Cardiovascular Center, Yokosuka Kyosai Hospital, Yokosuka, Japan
| | - Yuya Utsugi
- 1 Cardiovascular Center, Yokosuka Kyosai Hospital, Yokosuka, Japan
| | - Yuta Sudo
- 1 Cardiovascular Center, Yokosuka Kyosai Hospital, Yokosuka, Japan
| | - Shigeki Kimura
- 1 Cardiovascular Center, Yokosuka Kyosai Hospital, Yokosuka, Japan
| | - Takashi Ashikaga
- 2 Cardiovascular Medicine, Tokyo Medical and Dental University, Tokyo, Japan
| | | | - Mitsuaki Isobe
- 2 Cardiovascular Medicine, Tokyo Medical and Dental University, Tokyo, Japan
| |
Collapse
|
35
|
Boffa MB. Emerging Therapeutic Options for Lowering of Lipoprotein(a): Implications for Prevention of Cardiovascular Disease. Curr Atheroscler Rep 2017; 18:69. [PMID: 27761705 DOI: 10.1007/s11883-016-0622-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
PURPOSE OF REVIEW Elevated plasma concentrations of lipoprotein(a) (Lp(a)) are an independent and causal risk factor for cardiovascular diseases including coronary artery disease, ischemic stroke, and calcific aortic valve stenosis. This review summarizes the rationale for Lp(a) lowering and surveys relevant clinical trial data using a variety of agents capable of lowering Lp(a). RECENT FINDINGS Contemporary guidelines and recommendations outline populations of patients who should be screened for elevated Lp(a) and who might benefit from Lp(a) lowering. Therapies including drugs and apheresis have been described that lower Lp(a) levels modestly (∼20 %) to dramatically (∼80 %). Existing therapies that lower Lp(a) also have beneficial effects on other aspects of the lipid profile, with the exception of Lp(a)-specific apheresis and an antisense oligonucleotide that targets the mRNA encoding apolipoprotein(a). No clinical trials conducted to date have managed to answer the key question of whether Lp(a) lowering confers a benefit in terms of ameliorating cardiovascular risk, although additional outcome trials of therapies that lower Lp(a) are ongoing. It is more likely, however, that Lp(a)-specific agents will provide the most appropriate approach for addressing this question.
Collapse
Affiliation(s)
- Michael B Boffa
- Department of Biochemistry, Room 4245A Robarts Research Institute, University of Western Ontario, 1151 Richmond Street North, London, ON, Canada, N6A 5B7.
| |
Collapse
|
36
|
Roles of the low density lipoprotein receptor and related receptors in inhibition of lipoprotein(a) internalization by proprotein convertase subtilisin/kexin type 9. PLoS One 2017; 12:e0180869. [PMID: 28750079 PMCID: PMC5531514 DOI: 10.1371/journal.pone.0180869] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2016] [Accepted: 06/22/2017] [Indexed: 12/19/2022] Open
Abstract
Elevated plasma concentrations of lipoprotein(a) (Lp(a)) are a causal risk factor for cardiovascular disease. The mechanisms underlying Lp(a) clearance from plasma remain unclear, which is an obvious barrier to the development of therapies to specifically lower levels of this lipoprotein. Recently, it has been documented that monoclonal antibody inhibitors of proprotein convertase subtilisin/kexin type 9 (PCSK9) can lower plasma Lp(a) levels by 30%. Since PCSK9 acts primarily through the low density lipoprotein receptor (LDLR), this result is in conflict with the prevailing view that the LDLR does not participate in Lp(a) clearance. To support our recent findings in HepG2 cells that the LDLR can act as a bona fide receptor for Lp(a) whose effects are sensitive to PCSK9, we undertook a series of Lp(a) internalization experiments using different hepatic cells, with different variants of PCSK9, and with different members of the LDLR family. We found that PCSK9 decreased Lp(a) and/or apo(a) internalization by Huh7 human hepatoma cells and by primary mouse and human hepatocytes. Overexpression of human LDLR appeared to enhance apo(a)/Lp(a) internalization in both types of primary cells. Importantly, internalization of Lp(a) by LDLR-deficient mouse hepatocytes was not affected by PCSK9, but the effect of PCSK9 was restored upon overexpression of human LDLR. In HepG2 cells, Lp(a) internalization was decreased by gain-of-function mutants of PCSK9 more than by wild-type PCSK9, and a loss-of function variant had a reduced ability to influence Lp(a) internalization. Apo(a) internalization by HepG2 cells was not affected by apo(a) isoform size. Finally, we showed that very low density lipoprotein receptor (VLDLR), LDR-related protein (LRP)-8, and LRP-1 do not play a role in Lp(a) internalization or the effect of PCSK9 on Lp(a) internalization. Our findings are consistent with the idea that PCSK9 inhibits Lp(a) clearance through the LDLR, but do not exclude other effects of PCSK9 such as on Lp(a) biosynthesis.
Collapse
|
37
|
Ferretti G, Bacchetti T, Johnston TP, Banach M, Pirro M, Sahebkar A. Lipoprotein(a): A missing culprit in the management of athero-thrombosis? J Cell Physiol 2017; 233:2966-2981. [DOI: 10.1002/jcp.26050] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2017] [Accepted: 06/12/2017] [Indexed: 12/24/2022]
Affiliation(s)
- Gianna Ferretti
- Department of Clinical Sciences (DISCO); Polytechnic University of Marche; Marche Italy
| | - Tiziana Bacchetti
- Department of Life and Environmental Sciences (DISVA); Polytechnic University of Marche; Marche Italy
| | - Thomas P. Johnston
- Division of Pharmaceutical Sciences; School of Pharmacy; University of Missouri-Kansas City; Kansas City Missouri
| | - Maciej Banach
- Department of Hypertension; WAM University Hospital in Lodz; Medical University of Lodz; Lodz Poland
- Polish Mother's Memorial Hospital Research Institute (PMMHRI); Lodz Poland
| | - Matteo Pirro
- Unit of Internal Medicine; Angiology and Arteriosclerosis Diseases; Department of Medicine; University of Perugia; Perugia Italy
| | - Amirhossein Sahebkar
- Biotechnology Research Center; Mashhad University of Medical Sciences; Mashhad Iran
- Neurogenic Inflammation Research Center; Mashhad University of Medical Sciences; Mashhad Iran
| |
Collapse
|
38
|
Takahashi S. Triglyceride Rich Lipoprotein -LPL-VLDL Receptor and Lp(a)-VLDL Receptor Pathways for Macrophage Foam Cell Formation. J Atheroscler Thromb 2017; 24:552-559. [PMID: 28428482 PMCID: PMC5453679 DOI: 10.5551/jat.rv17004] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Very low-density lipoprotein (VLDL) receptor is a member of the low-density lipoprotein (LDL) receptor family. It binds triglyceride rich lipoprotein (TGRL) but not LDL, because it recognizes apolipoprotein (apo)E only but not apoB. The VLDL receptor functions as a peripheral lipoprotein receptor in concert with lipoprotein lipase (LPL) in heart, muscle, adipose tissue and macrophages. In contrast to the LDL receptor, VLDL receptor binds apo E2/2 VLDL and apoE3/3 VLDL particles, and its expression is not down-regulated by intracellular lipoproteins. It has been reported that both LDL-cholesterol (LDL-C) and postprandial triglyceride (chyromicron and VLDL remnants) are risk factors for human atherosclerotic cardiovascular disease (ASCVD). True ligands such as lipoprotein particles of the VLDL receptor are chyromicron remnant (CMR) and VLDL remnant (postprandial hyperlipidemia). Although the oxidized LDL (oxLDL)-scavenger receptors pathway is considered to be the main mechanism for macrophage foam cell formation, it seems that the TGRL-LPL-VLDL receptor pathway is also involved. Since Lp(a) is one of the ligands for the VLDL receptor, the Lp(a)- VLDL receptor pathway is another potential alternative. The expression of VLDL receptor protein in mouse macrophages is modest compared to that in rabbit and human macrophages, both in vitro and in vivo. Therefore, we need to elucidate the mechanism of human ASCVD not by using the mouse model and scavenger receptors pathway but instead using the rabbit model and VLDL receptor pathway, respectively.
Collapse
|
39
|
Abstract
Ischemic heart disease is the main cause of death worldwide and it is accelerated by increased low-density lipoprotein (LDL) cholesterol (LDL-C) and/or lipoprotein (a) (Lp(a)) concentrations. Proprotein convertase subtilisin/kexin type 9 (PCSK9) alters both LDL-C and in part Lp(a) concentrations through its ability to induce degradation of the LDL receptor (LDLR). PCSK9, however, has additional targets which are potentially involved in lipid metabolism regulation such as the very low density lipoprotein receptor (VLDL), CD36 (cluster of differentiation 36) and the epithelial cholesterol transporter (NPC1L1) and it affects expression of apolipoprotein B48. The PCSK9 activity is tightly regulated at several levels by factors influencing its transcription, secretion, or by extracellular inactivation and clearance. Many comorbidities (kidney insufficiency, hypothyreoidism, hyperinsulinemia, inflammation) modify PCSK9 expression and release. Two humanized antibodies directed against extracellular PCSK9 received approval by the European and US authorities and additional PCSK9 directed therapeutics (such as silencing RNA) are already in clinical trials. Their results demonstrate a significant reduction in both LDL-C and Lp(a) concentrations – independent of the concomitant medication – and one of them reduced plaque size in high risk cardiovascular patients; results of two ongoing large clinical endpoints studies are awaited. In this review, we summarize and discuss the recent biological data on PCSK9, the regulation of PCSK9, and finally briefly summarize the data of recent clinical studies in the context of lipid metabolism.
Collapse
Affiliation(s)
- Rainer Schulz
- Department of Physiology, Justus-Liebig-Universität, Aulweg 129, 35392, Giessen, Germany.
| | - Klaus-Dieter Schlüter
- Department of Physiology, Justus-Liebig-Universität, Aulweg 129, 35392, Giessen, Germany
| |
Collapse
|
40
|
Alarcón García JC, Rodríguez Suárez S, Muñiz Grijalvo O, García Morillo S. Vascular lesions in patient with Takayasu arteritis and massive elevated lipoprotein(a) levels. Residual involvement or premature aterosclerosis? CLINICA E INVESTIGACION EN ARTERIOSCLEROSIS 2017; 29:98-102. [PMID: 28188021 DOI: 10.1016/j.arteri.2016.10.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/05/2016] [Revised: 10/22/2016] [Accepted: 10/28/2016] [Indexed: 02/05/2023]
Abstract
Lipoprotein (a) [Lp(a)] is a lipoprotein defined by presenting a specific apolipoprotein, ApoA, linked to the ApoB-100 by different types of chemical bonds, including a disulfide bridge. Despite their atherogenic mechanism is not fully understood, its importance has been demonstrated in the development of premature aterosclerosis. Multiple studies have shown its role as a cardiovascular risk factor associated with heart disease and stroke. We report the case of a patient with a diagnosis of Takayasu arteritis in which a massive elevation of Lp(a) was detected. We emphasize its diagnostic and therapeutic implications.
Collapse
Affiliation(s)
- José Carlos Alarcón García
- Unidad de Colagenosis y Enfermedades Minoritarias, Unidad Experimental de Riesgo Cardiovascular, Servicio de Medicina Interna, Hospital Universitario Virgen del Rocío, Sevilla, España.
| | - Santiago Rodríguez Suárez
- Unidad de Colagenosis y Enfermedades Minoritarias, Unidad Experimental de Riesgo Cardiovascular, Servicio de Medicina Interna, Hospital Universitario Virgen del Rocío, Sevilla, España
| | - Ovidio Muñiz Grijalvo
- Unidad de Colagenosis y Enfermedades Minoritarias, Unidad Experimental de Riesgo Cardiovascular, Servicio de Medicina Interna, Hospital Universitario Virgen del Rocío, Sevilla, España
| | - Salvador García Morillo
- Unidad de Colagenosis y Enfermedades Minoritarias, Unidad Experimental de Riesgo Cardiovascular, Servicio de Medicina Interna, Hospital Universitario Virgen del Rocío, Sevilla, España
| |
Collapse
|
41
|
Sharma M, Redpath GM, Williams MJA, McCormick SPA. Recycling of Apolipoprotein(a) After PlgRKT-Mediated Endocytosis of Lipoprotein(a). Circ Res 2016; 120:1091-1102. [PMID: 28003220 DOI: 10.1161/circresaha.116.310272] [Citation(s) in RCA: 61] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/07/2016] [Revised: 12/15/2016] [Accepted: 12/21/2016] [Indexed: 11/16/2022]
Abstract
RATIONALE Lipoprotein(a) [Lp(a)] is a low-density lipoprotein-like lipoprotein and important cardiovascular risk factor whose cognate receptor and intracellular fate remains unknown. OBJECTIVE Our study aimed to determine the intracellular trafficking pathway for Lp(a) and the receptor responsible for its uptake in liver cells. METHODS AND RESULTS Human hepatoma cells were treated with Lp(a) purified from human plasma and Lp(a) uptake studied using Western blot analysis and intracellular localization of Lp(a) by confocal microscopy. Lp(a) was maximally internalized by 2 hours and was detected by an antiapo(a) antibody to be localized to Rab5-positive early endosomes, the trans-Golgi network, and subsequently Rab11-positive recycling endosomes. In human hepatoma cells, the apo(a) component from the internalized Lp(a) was resecreted back into the cellular media, whereas the low-density lipoprotein component was localized to the lysosomal compartment. Lp(a) internalization was reduced 0.35-fold in HAP1 and 0.33-fold in human hepatoma cells in which the plasminogen receptor (KT) was knocked out. Conversely, Lp(a) internalization was enhanced 2-fold in HAP1 and 1.6-fold in human hepatoma cells in which plasminogen receptor (KT) was overexpressed, showing for the first time the role of a specific plasminogen receptor in Lp(a) uptake. CONCLUSIONS The novel findings that Lp(a) is internalized by the plasminogen receptor, plasminogen receptor (KT), and the apo(a) component is recycled may have important implications for the catabolism and function of Lp(a).
Collapse
Affiliation(s)
- Monika Sharma
- From the Department of Biochemistry, Otago School of Medical Sciences, University of Otago, Dunedin, New Zealand (M.S., G.M.R., S.P.A.M.); and Department of Medicine, Dunedin School of Medicine, University of Otago, New Zealand (M.J.A.W.)
| | - Gregory M Redpath
- From the Department of Biochemistry, Otago School of Medical Sciences, University of Otago, Dunedin, New Zealand (M.S., G.M.R., S.P.A.M.); and Department of Medicine, Dunedin School of Medicine, University of Otago, New Zealand (M.J.A.W.)
| | - Michael J A Williams
- From the Department of Biochemistry, Otago School of Medical Sciences, University of Otago, Dunedin, New Zealand (M.S., G.M.R., S.P.A.M.); and Department of Medicine, Dunedin School of Medicine, University of Otago, New Zealand (M.J.A.W.)
| | - Sally P A McCormick
- From the Department of Biochemistry, Otago School of Medical Sciences, University of Otago, Dunedin, New Zealand (M.S., G.M.R., S.P.A.M.); and Department of Medicine, Dunedin School of Medicine, University of Otago, New Zealand (M.J.A.W.).
| |
Collapse
|
42
|
Bucci M, Tana C, Giamberardino MA, Cipollone F. Lp(a) and cardiovascular risk: Investigating the hidden side of the moon. Nutr Metab Cardiovasc Dis 2016; 26:980-986. [PMID: 27514608 DOI: 10.1016/j.numecd.2016.07.004] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/29/2016] [Revised: 06/21/2016] [Accepted: 07/06/2016] [Indexed: 12/21/2022]
Abstract
AIMS This article reports current evidence on the association between Lp(a) and cardiovascular (CV) disease and on pathophysiological mechanisms. The available information on therapy for reduction of lipoprotein(a) is also discussed. DATA SYNTHESIS Although some evidence is conflicting, Lp(a) seems to increase CV risk through stimulation of platelet aggregation, inhibition of tissue factor pathway inhibitor, alteration of fibrin clot structure and promotion of endothelial dysfunction and phospholipid oxidation. Lp(a) 3.5-fold higher than normal increases the risk of coronary heart disease and general CV events, particularly in those with LDL cholesterol ≥ 130 mg/dl. High Lp(a) values represent also an independent risk factor for ischemic stroke (more relevant in young stroke patients), peripheral artery disease (PAD) and aortic and mitral stenosis. Furthermore, high Lp(a) levels seem to be associated with increased risk of cardiovascular events in patients with chronic kidney disease, particularly in those undergoing percutaneous coronary intervention. CONCLUSIONS Lipoprotein (a) (Lp[a]) seems to significantly influence the risk of cardiovascular events. The effects of statins and fibrates on Lp(a) are limited and extremely variable. Nicotinic acid was shown effective in reducing Lp(a) but, due to its side effects and serious adverse events during clinical trials, it is no longer considered a possible option for treatment. To date, the treatment of choice for high levels of Lp(a) in high CV risk patients is represented by LDL-Apheresis. Thanks to innovative technologies, new selectively inhibiting LPA drugs are being developed and tested.
Collapse
Affiliation(s)
- M Bucci
- Regional Center for the Study of Atherosclerosis, Hypertension and Dyslipidemia, "SS Annunziata" Hospital - ASL Chieti, Italy; Ce.S.I.-Met, "G. D'Annunzio" University of Chieti, Italy
| | - C Tana
- Internal Medicine Unit, Guastalla Hospital, AUSL Reggio Emilia, Italy
| | - M A Giamberardino
- Ce.S.I.-Met, "G. D'Annunzio" University of Chieti, Italy; Geriatrics Clinic, Department of Medicine and Science of Aging, "G. D'Annunzio" University of Chieti, Italy
| | - F Cipollone
- Regional Center for the Study of Atherosclerosis, Hypertension and Dyslipidemia, "SS Annunziata" Hospital - ASL Chieti, Italy; Ce.S.I.-Met, "G. D'Annunzio" University of Chieti, Italy; Geriatrics Clinic, Department of Medicine and Science of Aging, "G. D'Annunzio" University of Chieti, Italy.
| |
Collapse
|
43
|
Yang X, Sethi A, Yanek LR, Knapper C, Nordestgaard BG, Tybjærg-Hansen A, Becker DM, Mathias RA, Remaley AT, Becker LC. SCARB1 Gene Variants Are Associated With the Phenotype of Combined High High-Density Lipoprotein Cholesterol and High Lipoprotein (a). ACTA ACUST UNITED AC 2016; 9:408-418. [PMID: 27651445 DOI: 10.1161/circgenetics.116.001402] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2016] [Accepted: 08/20/2016] [Indexed: 02/03/2023]
Abstract
BACKGROUND SR-B1 (scavenger receptor class B type 1), encoded by the gene SCARB1, is a lipoprotein receptor that binds both high-density lipoprotein (HDL) and low-density lipoprotein. We reported that SR-B1 is also a receptor for lipoprotein (a) (Lp(a)), mediating cellular uptake of Lp(a) in vitro and promoting clearance of Lp(a) in vivo. Although genetic variants in SCARB1 are associated with variations in HDL level, no SCARB1 variants affecting Lp(a) have been reported. METHODS AND RESULTS In an index subject with high levels of HDL cholesterol and Lp(a), SCARB1 was sequenced and demonstrated a missense mutation resulting in an S129L substitution in exon 3. To follow up, 2 cohorts (GeneSTAR, the family-based Genetic Study of Atherosclerosis Risk [n=543], and CCHS, the population-based Copenhagen City Heart Study [n=5835]) were screened for combined HDL cholesterol and Lp(a) elevations. Subjects with the extreme phenotype (HDL >80 mg/dL and Lp(a) >100 nmol/L in GeneSTAR, n=8, and >100 mg/dL in CCHS, n=9) underwent sequencing of SCARB1 exons; 15 of 18 from the combined population demonstrated genetic variants, including rare or uncommon missense or splice site mutations in 9 and homozygous synonymous variants in 6. Functional studies with 4 of the SCARB1 variants (c.386C>T, c.631-14T>G, c.4G>A, and c.631-53mC>T & c.726+55mCG>CA) showed decreased receptor function in vitro. CONCLUSIONS Human SCARB1 gene variants are associated with a new lipid phenotype, characterized by high levels of both HDL cholesterol and Lp(a). SCARB1 exonic variants often result in diminished function of translated SR-B1 via reduced binding/intracellular transport of Lp(a).
Collapse
Affiliation(s)
- Xiaoping Yang
- From the Department of Medicine, Johns Hopkins Medical Institutions, Baltimore, MD (X.Y., L.R.Y., D.M.B., R.A.M., L.C.B.); Lipoprotein Metabolism Section, Pulmonary and Vascular Medicine Branch, NHLBI, National Institutes of Health, Bethesda, MD (A.S., C.K., A.T.R.); and Copenhagen University Hospital, Faculty of Health Sciences, University of Copenhagen, Denmark (B.G.N., A.T.-H.)
| | - Amar Sethi
- From the Department of Medicine, Johns Hopkins Medical Institutions, Baltimore, MD (X.Y., L.R.Y., D.M.B., R.A.M., L.C.B.); Lipoprotein Metabolism Section, Pulmonary and Vascular Medicine Branch, NHLBI, National Institutes of Health, Bethesda, MD (A.S., C.K., A.T.R.); and Copenhagen University Hospital, Faculty of Health Sciences, University of Copenhagen, Denmark (B.G.N., A.T.-H.)
| | - Lisa R Yanek
- From the Department of Medicine, Johns Hopkins Medical Institutions, Baltimore, MD (X.Y., L.R.Y., D.M.B., R.A.M., L.C.B.); Lipoprotein Metabolism Section, Pulmonary and Vascular Medicine Branch, NHLBI, National Institutes of Health, Bethesda, MD (A.S., C.K., A.T.R.); and Copenhagen University Hospital, Faculty of Health Sciences, University of Copenhagen, Denmark (B.G.N., A.T.-H.)
| | - Cathy Knapper
- From the Department of Medicine, Johns Hopkins Medical Institutions, Baltimore, MD (X.Y., L.R.Y., D.M.B., R.A.M., L.C.B.); Lipoprotein Metabolism Section, Pulmonary and Vascular Medicine Branch, NHLBI, National Institutes of Health, Bethesda, MD (A.S., C.K., A.T.R.); and Copenhagen University Hospital, Faculty of Health Sciences, University of Copenhagen, Denmark (B.G.N., A.T.-H.)
| | - Børge G Nordestgaard
- From the Department of Medicine, Johns Hopkins Medical Institutions, Baltimore, MD (X.Y., L.R.Y., D.M.B., R.A.M., L.C.B.); Lipoprotein Metabolism Section, Pulmonary and Vascular Medicine Branch, NHLBI, National Institutes of Health, Bethesda, MD (A.S., C.K., A.T.R.); and Copenhagen University Hospital, Faculty of Health Sciences, University of Copenhagen, Denmark (B.G.N., A.T.-H.)
| | - Anne Tybjærg-Hansen
- From the Department of Medicine, Johns Hopkins Medical Institutions, Baltimore, MD (X.Y., L.R.Y., D.M.B., R.A.M., L.C.B.); Lipoprotein Metabolism Section, Pulmonary and Vascular Medicine Branch, NHLBI, National Institutes of Health, Bethesda, MD (A.S., C.K., A.T.R.); and Copenhagen University Hospital, Faculty of Health Sciences, University of Copenhagen, Denmark (B.G.N., A.T.-H.)
| | - Diane M Becker
- From the Department of Medicine, Johns Hopkins Medical Institutions, Baltimore, MD (X.Y., L.R.Y., D.M.B., R.A.M., L.C.B.); Lipoprotein Metabolism Section, Pulmonary and Vascular Medicine Branch, NHLBI, National Institutes of Health, Bethesda, MD (A.S., C.K., A.T.R.); and Copenhagen University Hospital, Faculty of Health Sciences, University of Copenhagen, Denmark (B.G.N., A.T.-H.)
| | - Rasika A Mathias
- From the Department of Medicine, Johns Hopkins Medical Institutions, Baltimore, MD (X.Y., L.R.Y., D.M.B., R.A.M., L.C.B.); Lipoprotein Metabolism Section, Pulmonary and Vascular Medicine Branch, NHLBI, National Institutes of Health, Bethesda, MD (A.S., C.K., A.T.R.); and Copenhagen University Hospital, Faculty of Health Sciences, University of Copenhagen, Denmark (B.G.N., A.T.-H.)
| | - Alan T Remaley
- From the Department of Medicine, Johns Hopkins Medical Institutions, Baltimore, MD (X.Y., L.R.Y., D.M.B., R.A.M., L.C.B.); Lipoprotein Metabolism Section, Pulmonary and Vascular Medicine Branch, NHLBI, National Institutes of Health, Bethesda, MD (A.S., C.K., A.T.R.); and Copenhagen University Hospital, Faculty of Health Sciences, University of Copenhagen, Denmark (B.G.N., A.T.-H.)
| | - Lewis C Becker
- From the Department of Medicine, Johns Hopkins Medical Institutions, Baltimore, MD (X.Y., L.R.Y., D.M.B., R.A.M., L.C.B.); Lipoprotein Metabolism Section, Pulmonary and Vascular Medicine Branch, NHLBI, National Institutes of Health, Bethesda, MD (A.S., C.K., A.T.R.); and Copenhagen University Hospital, Faculty of Health Sciences, University of Copenhagen, Denmark (B.G.N., A.T.-H.).
| |
Collapse
|
44
|
Raal FJ, Giugliano RP, Sabatine MS, Koren MJ, Blom D, Seidah NG, Honarpour N, Lira A, Xue A, Chiruvolu P, Jackson S, Di M, Peach M, Somaratne R, Wasserman SM, Scott R, Stein EA. PCSK9 inhibition-mediated reduction in Lp(a) with evolocumab: an analysis of 10 clinical trials and the LDL receptor's role. J Lipid Res 2016; 57:1086-96. [PMID: 27102113 DOI: 10.1194/jlr.p065334] [Citation(s) in RCA: 161] [Impact Index Per Article: 20.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2016] [Indexed: 12/12/2022] Open
Abstract
Lipoprotein (a) [Lp(a)] is independently associated with CVD risk. Evolocumab, a monoclonal antibody (mAb) to proprotein convertase subtilisin/kexin type 9 (PCSK9), decreases Lp(a). The potential mechanisms were assessed. A pooled analysis of Lp(a) and LDL cholesterol (LDL-C) in 3,278 patients from 10 clinical trials (eight phase 2/3; two extensions) was conducted. Within each parent study, biweekly and monthly doses of evolocumab statistically significantly reduced Lp(a) at week 12 versus control (P < 0.001 within each study); pooled median (quartile 1, quartile 3) percent reductions were 24.7% (40.0, 3.6) and 21.7% (39.9, 4.2), respectively. Reductions were maintained through week 52 of the open-label extension, and correlated with LDL-C reductions [with and without correction for Lp(a)-cholesterol] at both time points (P < 0.0001). The effect of LDL and LDL receptor (LDLR) availability on Lp(a) cell-association was measured in HepG2 cells: cell-associated LDL fluorescence was reversed by unlabeled LDL and Lp(a). Lp(a) cell-association was reduced by coincubation with LDL and PCSK9 and reversed by adding PCSK9 mAb. These studies support that reductions in Lp(a) with PCSK9 inhibition are partly due to increased LDLR-mediated uptake. In most situations, Lp(a) appears to compete poorly with LDL for LDLR binding and internalization, but when LDLR expression is increased with evolocumab, particularly in the setting of low circulating LDL, Lp(a) is reduced.
Collapse
Affiliation(s)
- Frederick J Raal
- Carbohydrate and Lipid Metabolism Research Unit, Faculty of Health Sciences, University of Witwatersrand, Johannesburg, South Africa
| | - Robert P Giugliano
- TIMI Study Group, Cardiovascular Medicine, Brigham and Women's Hospital, Boston, MA
| | - Marc S Sabatine
- TIMI Study Group, Cardiovascular Medicine, Brigham and Women's Hospital, Boston, MA
| | | | - Dirk Blom
- Division of Lipidology, University of Cape Town, Cape Town, South Africa
| | | | | | | | | | | | | | - Mei Di
- Amgen Inc., San Francisco, CA
| | | | | | | | | | - Evan A Stein
- Metabolic and Atherosclerosis Research Center, Cincinnati, OH
| |
Collapse
|
45
|
Abstract
Lipoprotein (a) [Lp(a)] is a modified LDL particle with an additional apolipoprotein [apo(a)] protein covalently attached by a thioester bond. Multiple isoforms of apo(a) exist that are genetically determined by differences in the number of Kringle-IV type-2 repeats encoded by the LPA gene. Elevated plasma Lp(a) is an independent risk factor for cardiovascular disease. The phenotypic diversity of familial Lp(a) hyperlipidemia [Lp(a)-HLP] and familial hypercholesterolemia [FH], as defined risks with genetic background, and their frequent co-incidence with additional cardiovascular risk factors require a critical revision of the current diagnostic and therapeutic recommendations established for isolated familial Lp(a)-HLP or FH in combination with elevated Lp(a) levels. Lp(a) assays still suffer from poor standardization, comparability and particle variation. Further evaluation of the current biomarkers and establishment of novel comorbidity biomarkers are necessary for extended risk assessment of cardiovascular disease in FH or Lp(a)-HLP and to better understand the pathophysiology and to improve patient stratification of the Lp(a) syndrome complex. Lp(a) promotes vascular remodeling, increased lesion progression and intima media thickening through induction of M1-macrophages, antiangiogenic effects (e.g. vasa vasorum) with secretion of the antiangiogenic chemokine CXCL10 (IP10) and CXCR3 mediated activation of Th1- and NK-cells. In addition inhibition of serine proteases causing disturbances of thrombosis/ hemostasis/ fibrinolysis, TGFb-activation and acute phase response (e.g. CRP, anti-PL antibodies) are major features of Lp(a) pathology. Anti-PL antibodies (EO6 epitope) also bind to oxidized Lp(a). Lipoprotein apheresis is used to reduce circulating lipoproteins in patients with severe FH and/or Lp(a)-HLP, particularly with multiple cardiovascular risks who are intolerant or insufficiently responsive to lipid-lowering drugs.
Collapse
|
46
|
Kazemian MR, Solouk A, Tan A, Seifalian AM. Preventing in-stent restenosis using lipoprotein (a), lipid and cholesterol adsorbent materials. Med Hypotheses 2015; 85:986-8. [DOI: 10.1016/j.mehy.2015.08.023] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2015] [Accepted: 08/31/2015] [Indexed: 11/16/2022]
|
47
|
Damsteegt EL, Mizuta H, Hiramatsu N, Lokman PM. How do eggs get fat? Insights into ovarian fatty acid accumulation in the shortfinned eel, Anguilla australis. Gen Comp Endocrinol 2015; 221:94-100. [PMID: 25660471 DOI: 10.1016/j.ygcen.2014.12.019] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2014] [Revised: 12/17/2014] [Accepted: 12/31/2014] [Indexed: 11/30/2022]
Abstract
Previous research using eels has shown that 11-ketotestosterone can induce ovarian triacylglyceride accumulation both in vivo and in vitro. Further, accumulation is dramatically enhanced in the presence of very-low density lipoprotein. This study examined the involvement of the low density lipoprotein receptor and vitellogenin receptor in oocyte lipid accumulation. Specific antisera were used in an attempt to block the vitellogenin receptor and/or the low density lipoprotein receptor. Accordingly, incubation with the low density lipoprotein receptor antiserum clearly reduced the oocyte diameter and the amount of oil present within the oocyte. In contrast, blocking the vitellogenin receptor had little effect on either oocyte surface area or the abundance of oil droplets in the cytosol. In keeping with birds, we conclude that the low density lipoprotein receptor is a major player involved in mediating ovarian fatty acid accumulation in the eel. However, lipoprotein lipase-mediated fatty acid accumulation also remains conceivable, for example through interactions between this enzyme and the low density lipoprotein receptor.
Collapse
Affiliation(s)
- Erin L Damsteegt
- Department of Zoology, University of Otago, 340 Great King Street, PO Box 56, Dunedin 9054, New Zealand.
| | - Hiroko Mizuta
- Faculty of Fisheries Sciences, Hokkaido University, 3-1-1 Minato-cho, Hakodate, Hokkaido 041-8611, Japan
| | - Naoshi Hiramatsu
- Faculty of Fisheries Sciences, Hokkaido University, 3-1-1 Minato-cho, Hakodate, Hokkaido 041-8611, Japan
| | - P Mark Lokman
- Department of Zoology, University of Otago, 340 Great King Street, PO Box 56, Dunedin 9054, New Zealand
| |
Collapse
|
48
|
Yeang C, Tsimikas S. HDL-C, ABCA1-mediated cholesterol efflux, and lipoprotein(a): insights into a potential novel physiologic role of lipoprotein(a). J Lipid Res 2015; 56:1241-4. [PMID: 26014961 DOI: 10.1194/jlr.e060947] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Affiliation(s)
- Calvin Yeang
- Division of Cardiovascular Medicine, University of California San Diego, Sulpizio Cardiovascular Center, La Jolla, CA
| | - Sotirios Tsimikas
- Division of Cardiovascular Medicine, University of California San Diego, Sulpizio Cardiovascular Center, La Jolla, CA
| |
Collapse
|
49
|
Sharma M, Von Zychlinski-Kleffmann A, Porteous CM, Jones GT, Williams MJA, McCormick SPA. Lipoprotein (a) upregulates ABCA1 in liver cells via scavenger receptor-B1 through its oxidized phospholipids. J Lipid Res 2015; 56:1318-28. [PMID: 25852127 PMCID: PMC4479336 DOI: 10.1194/jlr.m056150] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2015] [Indexed: 01/04/2023] Open
Abstract
Elevated levels of lipoprotein (a) [Lp(a)] are a well-established risk factor for developing CVD. While Lp(a) levels are thought to be independent of other plasma lipoproteins, some trials have reported a positive association between Lp(a) and HDL. Whether Lp(a) has a direct effect on HDL is not known. Here we investigated to determine whether Lp(a) had any effect on the ABCA1 pathway of HDL production in liver cells. Incubation of HepG2 cells with Lp(a) upregulated the PPARγ protein by 1.7-fold and the liver X receptor α protein by 3-fold. This was accompanied by a 1.8-fold increase in ABCA1 protein and a 1.5-fold increase in cholesterol efflux onto apoA1. We showed that Lp(a) was internalized by HepG2 cells, however, the ABCA1 response to Lp(a) was mediated by the selective uptake of oxidized phospholipids (oxPLs) from Lp(a) via the scavenger receptor-B1 and not by Lp(a) internalization per se. We conclude that there is a biological connection between Lp(a) and HDL through the ability of Lp(a)’s oxPLs to upregulate HDL biosynthesis.
Collapse
Affiliation(s)
- Monika Sharma
- Department of Biochemistry, University of Otago, Dunedin, New Zealand
| | | | | | - Gregory T Jones
- Departments of Surgical Sciences University of Otago, Dunedin, New Zealand
| | | | | |
Collapse
|
50
|
Maranhão RC, Carvalho PO, Strunz CC, Pileggi F. Lipoprotein (a): structure, pathophysiology and clinical implications. Arq Bras Cardiol 2015; 103:76-84. [PMID: 25120086 PMCID: PMC4126764 DOI: 10.5935/abc.20140101] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2013] [Accepted: 10/01/2013] [Indexed: 11/20/2022] Open
Abstract
The chemical structure of lipoprotein (a) is similar to that of LDL, from which it
differs due to the presence of apolipoprotein (a) bound to apo B100 via one disulfide
bridge. Lipoprotein (a) is synthesized in the liver and its plasma concentration,
which can be determined by use of monoclonal antibody-based methods, ranges from <
1 mg to > 1,000 mg/dL. Lipoprotein (a) levels over 20-30 mg/dL are associated with
a two-fold risk of developing coronary artery disease. Usually, black subjects have
higher lipoprotein (a) levels that, differently from Caucasians and Orientals, are
not related to coronary artery disease. However, the risk of black subjects must be
considered. Sex and age have little influence on lipoprotein (a) levels. Lipoprotein
(a) homology with plasminogen might lead to interference with the fibrinolytic
cascade, accounting for an atherogenic mechanism of that lipoprotein. Nevertheless,
direct deposition of lipoprotein (a) on arterial wall is also a possible mechanism,
lipoprotein (a) being more prone to oxidation than LDL. Most prospective studies have
confirmed lipoprotein (a) as a predisposing factor to atherosclerosis. Statin
treatment does not lower lipoprotein (a) levels, differently from niacin and
ezetimibe, which tend to reduce lipoprotein (a), although confirmation of ezetimibe
effects is pending. The reduction in lipoprotein (a) concentrations has not been
demonstrated to reduce the risk for coronary artery disease. Whenever higher
lipoprotein (a) concentrations are found, and in the absence of more effective and
well-tolerated drugs, a more strict and vigorous control of the other coronary artery
disease risk factors should be sought.
Collapse
Affiliation(s)
- Raul Cavalcante Maranhão
- Instituto do Coração, Hospital das Clínicas, Faculdade de Medicina, Universidade de São Paulo, São Paulo, SP, Brazil
| | - Priscila Oliveira Carvalho
- Instituto do Coração, Hospital das Clínicas, Faculdade de Medicina, Universidade de São Paulo, São Paulo, SP, Brazil
| | - Celia Cassaro Strunz
- Instituto do Coração, Hospital das Clínicas, Faculdade de Medicina, Universidade de São Paulo, São Paulo, SP, Brazil
| | - Fulvio Pileggi
- Instituto do Coração, Hospital das Clínicas, Faculdade de Medicina, Universidade de São Paulo, São Paulo, SP, Brazil
| |
Collapse
|