1
|
Cao Zhang AM, Ziogos E, Harb T, Gerstenblith G, Leucker TM. Emerging clinical role of proprotein convertase subtilisin/kexin type 9 inhibition-Part one: Pleiotropic pro-atherosclerotic effects of PCSK9. Eur J Clin Invest 2024; 54:e14273. [PMID: 38922860 DOI: 10.1111/eci.14273] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Revised: 05/20/2024] [Accepted: 06/10/2024] [Indexed: 06/28/2024]
Abstract
BACKGROUND Proprotein convertase subtilisin/kexin type 9 (PCSK9) is primarily recognized for its role in lipid metabolism, but recent evidence suggests that it may have broader implications due to its diverse tissue expression. OBJECTIVE This review aims to explore the multifaceted functions of PCSK9, highlighting its pro-atherosclerotic effects, including its impact on circulating lipoprotein variables, non-low-density lipoprotein receptors, and various cell types involved in atherosclerotic plaque development. CONCLUSIONS PCSK9 exhibits diverse roles beyond lipid metabolism, potentially contributing to atherosclerosis through multiple pathways. Understanding these mechanisms could offer new insights into therapeutic strategies targeting PCSK9 for cardiovascular disease management.
Collapse
Affiliation(s)
- Alexander M Cao Zhang
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Efthymios Ziogos
- Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Tarek Harb
- Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Gary Gerstenblith
- Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Thorsten M Leucker
- Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
2
|
Li B, Liu Y, Zhou X, Gu W, Mu Y. Remnant cholesterol, but not other traditional lipids or lipid ratios, is independently and positively related to future diabetes risk in Chinese general population: A 3 year cohort study. J Diabetes Investig 2024; 15:1084-1093. [PMID: 38568177 PMCID: PMC11292390 DOI: 10.1111/jdi.14205] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/01/2024] [Revised: 03/05/2024] [Accepted: 03/19/2024] [Indexed: 08/02/2024] Open
Abstract
AIMS Very few cohort studies are available about the relation between remnant cholesterol (RC) and diabetes. Based on a prospective cohort survey, this research aimed at investigating if high RC was related to a future diabetes risk in the Chinese population, as well as to compare the association between RC, high-density lipoprotein cholesterol (HDL-C), low-density lipoprotein cholesterol (LDL-C), total cholesterol (TC), triglycerides (TG), TG/HDL-C, LDL-C/HDL-C, TC/HDL-C, and non-high-density lipoprotein cholesterol (non-HDL-C), and future diabetes risk. MATERIALS AND METHODS 6,700 baseline normoglycemic participants of the REACTION study's Beijing center were recruited in 2011-2012 and followed up in 2015. Multivariate Cox regression analyses were performed to explore the relationship of RC, HDL-C, LDL-C, TC, TG, LDL-C/HDL-C, TG/HDL-C, TC/HDL-C, and non-HDL-C and a future diabetes risk. RESULTS After potential confounders were adjusted for, only RC (HR 1.134, 95% CI 1.016-1.267, P = 0.025) was positively related to a future diabetes risk, and only HDL-C (HR 0.728, 95% CI 0.578-0.918, P = 0.007) was negatively related to a future diabetes risk. The rest of the lipid parameters were not related to a future risk of diabetes. Sensitivity and stratification analyses revealed that the relation between RC and future diabetes risk was stable. RC and future diabetes risk were still positively correlated even when the HDL-C was ≥1.04 mmol/L (HR 1.167, 95% CI 1.050-1.297, P = 0.004). CONCLUSIONS It was RC, but not other lipid parameters, that was independently and positively related to a future risk of diabetes among the Chinese general population. Moreover, the relationship between RC and diabetes risk was stable, even with appropriate levels of HDL-C.
Collapse
Affiliation(s)
- Binqi Li
- Medical CollegeNankai UniversityTianjinChina
- Department of Endocrinology, First Medical CenterChinese PLA General HospitalBeijingChina
| | - Yang Liu
- Department of Endocrinology, First Medical CenterChinese PLA General HospitalBeijingChina
- Department of Endocrinology, Eighth Medical CenterChinese PLA General HospitalBeijingChina
| | - Xin Zhou
- School of Graduate StudiesChinese PLA General HospitalBeijingChina
- Second Medical CenterChinese PLA General HospitalBeijingChina
| | - Weijun Gu
- Department of Endocrinology, First Medical CenterChinese PLA General HospitalBeijingChina
- School of Graduate StudiesChinese PLA General HospitalBeijingChina
| | - Yiming Mu
- Medical CollegeNankai UniversityTianjinChina
| |
Collapse
|
3
|
Li B, Liu Y, Zhou X, Chen L, Yan L, Tang X, Gao Z, Wan Q, Luo Z, Qin G, Ning G, Gu W, Mu Y. Remnant cholesterol is more positively related to diabetes, prediabetes, and insulin resistance than conventional lipid parameters and lipid ratios: A multicenter, large sample survey. J Diabetes 2024; 16:e13592. [PMID: 39136535 PMCID: PMC11320755 DOI: 10.1111/1753-0407.13592] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Revised: 04/30/2024] [Accepted: 05/17/2024] [Indexed: 08/16/2024] Open
Abstract
BACKGROUND Not many large-sample investigations are available that compare the potency of the relationship of remnant cholesterol (RC) and other lipid parameters with diabetes and prediabetes. The goals of our study are to discover the relationship between RC and prediabetes, diabetes, and insulin resistance (IR) and to investigate RC, high-density lipoprotein cholesterol (HDL-C), non-HDL-C, triglycerides (TG), low-density lipoprotein cholesterol (LDL-C), total cholesterol (TC), TC/HDL-C, LDL-C/HDL-C, and TG/HDL-C, which are the lipid parameters that are most positively related to diabetes, prediabetes, and IR. METHODS This research enrolled 36 684 subjects from China's eight provinces. We employed multiple logistic regression analysis for testing the relationship between lipid parameters and diabetes, prediabetes, and IR. RESULTS After adjusting for potential confounders, and comparing the results with other lipid parameters, the positive relationship between RC and diabetes (odds ratio [OR] 1.417, 95% confidence interval [CI]: 1.345-1.492), prediabetes (OR 1.555, 95% CI: 1.438-1.628), and IR (OR 1.488, 95% CI: 1.404-1.577) was highest. RC was still related to diabetes, prediabetes, and IR even when TG <2.3 mmol/L (diabetes: OR 1.256, 95% CI: 1.135-1.390; prediabetes: OR 1.503, 95% CI: 1.342-1.684; and IR: OR 1.278, 95% CI: 1.140-1.433), LDL-C <2.6 mmol/L (diabetes: OR 1.306, 95% CI: 1.203-1.418; prediabetes: OR 1.597, 95% CI: 1.418-1.798; and IR: OR 1.552, 95% CI: 1.416-1.701), or HDL-C ≥1 mmol/L (diabetes: OR 1.456, 95% CI: 1.366-1.550; prediabetes: OR 1.553, 95% CI: 1.421-1.697; and IR: OR 1.490, 95% CI: 1.389-1.598). CONCLUSION RC is more positively related to diabetes, prediabetes, and IR than conventional lipids and lipid ratios in the general population, the relationships between RC and diabetes, prediabetes, and IR are stable, even if HDL-C, LDL-C, or TG are at appropriate levels.
Collapse
Affiliation(s)
- Binqi Li
- School of MedicineNankai UniversityTianjinChina
- Department of EndocrinologyFirst Medical Center of PLA General HospitalBeijingChina
| | - Yang Liu
- Department of EndocrinologyFirst Medical Center of PLA General HospitalBeijingChina
- Department of EndocrinologyEighth Medical Center of PLA General HospitalBeijingChina
| | - Xin Zhou
- Graduate SchoolChinese PLA General HospitalBeijingChina
- Department of Medical OncologyFifth Medical Center of Chinese PLA General HospitalBeijingChina
- The Second Medical Center of Chinese PLA General HospitalBeijingChina
| | - Lulu Chen
- Wuhan Union HospitalHuazhong University of Science and TechnologyWuhanChina
| | - Li Yan
- Department of EndocrinologyZhongshan University Sun Yat‐sen Memorial HospitalGuangzhouChina
| | - Xulei Tang
- Department of EndocrinologyFirst Hospital of Lanzhou UniversityLanzhouChina
| | - Zhengnan Gao
- Department of EndocrinologyDalian Central HospitalDalianChina
| | - Qin Wan
- Department of EndocrinologySouthwest Medical University Affiliated HospitalLuzhouChina
| | - Zuojie Luo
- Department of endocrinologyFirst Affiliated Hospital of Guangxi Medical UniversityNanningChina
| | - Guijun Qin
- Department of endocrinologyFirst Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| | - Guang Ning
- Department of Endocrinology, Ruijin HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Weijun Gu
- Department of EndocrinologyFirst Medical Center of PLA General HospitalBeijingChina
- Department of EndocrinologyEighth Medical Center of PLA General HospitalBeijingChina
- Graduate SchoolChinese PLA General HospitalBeijingChina
| | - Yiming Mu
- School of MedicineNankai UniversityTianjinChina
- Department of EndocrinologyFirst Medical Center of PLA General HospitalBeijingChina
- Department of EndocrinologyEighth Medical Center of PLA General HospitalBeijingChina
- Graduate SchoolChinese PLA General HospitalBeijingChina
| |
Collapse
|
4
|
Xu M, Zhang P, Lv W, Chen Y, Chen M, Leng Y, Hu T, Wang K, Zhao Y, Shen J, You X, Gu D, Zhao W, Tan S. A bifunctional anti-PCSK9 scFv/Exendin-4 fusion protein exhibits enhanced lipid-lowering effects via targeting multiple signaling pathways in HFD-fed mice. Int J Biol Macromol 2023; 253:127003. [PMID: 37739280 DOI: 10.1016/j.ijbiomac.2023.127003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Revised: 05/14/2023] [Accepted: 09/17/2023] [Indexed: 09/24/2023]
Abstract
Fusion protein which encompasses more than one functional component, has become one of the most important representatives of macromolecular drugs for disease treatment since that monotherapy itself might not be effective enough to eradicate the disease. In this study, we sought to construct a bifunctional antibody fusion protein by fusing anti-PCSK9 scFv with Exendin-4 for simultaneously lowering both LDL-C and TG. Firstly, three Ex4-anti-PCSK9 scFv fusion proteins were constructed by genetically connecting the C-terminal of Exendin-4 to the N-terminal of anti-PCSK9 scFv through various flexible linker peptides (G4S)n (n = 2, 3, 4). After soluble expression in E. coli, the most potent Ex4-(G4S)4-anti-PCSK9 scFv fusion protein was selected based on in vitro activity assays. Then, we investigated the in vivo therapeutic effects of Ex4-(G4S)4-anti-PCSK9 scFv on the serum lipid profile and bodyweight changes as well as underlying molecular mechanism in HFD-fed C57BL/6 mice. The data showed that Ex4-(G4S)4-anti-PCSK9 scFv exhibits enhanced effects of lowering both LDL-C and TG in serum, reducing food intake and body weight via blocking PCSK9/LDLR, activating AMPK/SREBP-1 pathways, and up-regulating sirt6. Conclusively, Ex4-(G4S)4-anti-PCSK9 has the potential to serve as a promising therapeutic agent for effectively treating dyslipidemia with high levels of both LDL-C and TG.
Collapse
Affiliation(s)
- Menglong Xu
- Department of Cell and Molecular Biology, School of Life Science and Technology, State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Druggability of Biopharmaceuticals, China Pharmaceutical University, Nanjing 210009, PR China
| | - Panpan Zhang
- Department of Cell and Molecular Biology, School of Life Science and Technology, State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Druggability of Biopharmaceuticals, China Pharmaceutical University, Nanjing 210009, PR China
| | - Wenxiu Lv
- Department of Cell and Molecular Biology, School of Life Science and Technology, State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Druggability of Biopharmaceuticals, China Pharmaceutical University, Nanjing 210009, PR China
| | - Yuting Chen
- Department of Cell and Molecular Biology, School of Life Science and Technology, State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Druggability of Biopharmaceuticals, China Pharmaceutical University, Nanjing 210009, PR China
| | - Manman Chen
- Department of Cell and Molecular Biology, School of Life Science and Technology, State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Druggability of Biopharmaceuticals, China Pharmaceutical University, Nanjing 210009, PR China
| | - Yeqing Leng
- Department of Cell and Molecular Biology, School of Life Science and Technology, State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Druggability of Biopharmaceuticals, China Pharmaceutical University, Nanjing 210009, PR China
| | - Tuo Hu
- Department of Cell and Molecular Biology, School of Life Science and Technology, State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Druggability of Biopharmaceuticals, China Pharmaceutical University, Nanjing 210009, PR China
| | - Ke Wang
- Department of Cell and Molecular Biology, School of Life Science and Technology, State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Druggability of Biopharmaceuticals, China Pharmaceutical University, Nanjing 210009, PR China
| | - Yaqiang Zhao
- Department of Cell and Molecular Biology, School of Life Science and Technology, State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Druggability of Biopharmaceuticals, China Pharmaceutical University, Nanjing 210009, PR China
| | - Jiaqi Shen
- Department of Cell and Molecular Biology, School of Life Science and Technology, State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Druggability of Biopharmaceuticals, China Pharmaceutical University, Nanjing 210009, PR China
| | - Xiangyan You
- Department of Cell and Molecular Biology, School of Life Science and Technology, State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Druggability of Biopharmaceuticals, China Pharmaceutical University, Nanjing 210009, PR China
| | - Dian Gu
- Department of Cell and Molecular Biology, School of Life Science and Technology, State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Druggability of Biopharmaceuticals, China Pharmaceutical University, Nanjing 210009, PR China
| | - Wenfeng Zhao
- Department of Cell and Molecular Biology, School of Life Science and Technology, State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Druggability of Biopharmaceuticals, China Pharmaceutical University, Nanjing 210009, PR China
| | - Shuhua Tan
- Department of Cell and Molecular Biology, School of Life Science and Technology, State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Druggability of Biopharmaceuticals, China Pharmaceutical University, Nanjing 210009, PR China.
| |
Collapse
|
5
|
Yuan L, Liu J, Huang Z, Zhao Y, Feng Y, Yang X, Hu H, Zhang J, Li T, Li Y, Wu Y, Li X, Huang H, Hu F, Hu D, Zhang M. Elevated remnant cholesterol increase 6-year type 2 diabetes mellitus onset risk. Clin Chim Acta 2023; 541:117253. [PMID: 36796575 DOI: 10.1016/j.cca.2023.117253] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Revised: 02/06/2023] [Accepted: 02/10/2023] [Indexed: 02/16/2023]
Abstract
AIM To investigate the association of remnant cholesterol (RC) with future type 2 diabetes mellitus (T2DM) risk, and to assess the underlying impact of some recognized risk factors on it. METHODS A total of 11468 nondiabetic adults in rural China were recruited in 2007-2008 and followed up in 2013-2014. Logistic regression was used to assess the risk of incident T2DM by quartiles of baseline RC, estimating odds ratios (ORs) and 95% confidence intervals (CIs). Association of the combinations of RC and low-density lipoprotein cholesterol (LDL-C) with T2DM risk were further evaluated. RESULTS Multivariable adjusted OR (95% CI) for incident T2DM associated with quartile 4 versus quartile 1 of RC was 2.72 (2.05-3.62). Per 1-standard deviation (SD) increases in RC levels was associated with a 34% higher T2DM risk. However, gender modified the specific association (Pinteraction < 0.05), with the association stronger among females. When combining the low LDL-C and low RC groups as reference, individuals with RC levels ≥ 0.56 mmol/L had more than a 2-fold T2DM risk, regardless of LDL-C level. CONCLUSIONS Elevated RC levels increase T2DM risk in rural Chinese populations. In those who cannot control their risk by lowering LDL-C levels, the goal of lipid-lowering therapy can be shifted to RC.
Collapse
Affiliation(s)
- Lijun Yuan
- Department of General Practice, The Affiliated Luohu Hospital of Shenzhen University Medical School, Shenzhen, Guangdong, People's Republic of China
| | - Jiong Liu
- Department of General Practice, The Affiliated Luohu Hospital of Shenzhen University Medical School, Shenzhen, Guangdong, People's Republic of China; Department of Biostatistics and Epidemiology, School of Public Health, Shenzhen University Medical School, Shenzhen, Guangdong, People's Republic of China
| | - Zelin Huang
- Department of General Practice, The Affiliated Luohu Hospital of Shenzhen University Medical School, Shenzhen, Guangdong, People's Republic of China; Department of Biostatistics and Epidemiology, School of Public Health, Shenzhen University Medical School, Shenzhen, Guangdong, People's Republic of China
| | - Yang Zhao
- Department of Epidemiology and Health Statistics, College of Public Health, Zhengzhou University, Zhengzhou, Henan, People's Republic of China
| | - Yifei Feng
- Department of Epidemiology and Health Statistics, College of Public Health, Zhengzhou University, Zhengzhou, Henan, People's Republic of China
| | - Xingjin Yang
- Department of Epidemiology and Health Statistics, College of Public Health, Zhengzhou University, Zhengzhou, Henan, People's Republic of China
| | - Huifang Hu
- Department of Epidemiology and Health Statistics, College of Public Health, Zhengzhou University, Zhengzhou, Henan, People's Republic of China
| | - Jinli Zhang
- Department of Epidemiology and Health Statistics, College of Public Health, Zhengzhou University, Zhengzhou, Henan, People's Republic of China
| | - Tianze Li
- Department of Epidemiology and Health Statistics, College of Public Health, Zhengzhou University, Zhengzhou, Henan, People's Republic of China
| | - Yang Li
- Department of General Practice, The Affiliated Luohu Hospital of Shenzhen University Medical School, Shenzhen, Guangdong, People's Republic of China; Department of Biostatistics and Epidemiology, School of Public Health, Shenzhen University Medical School, Shenzhen, Guangdong, People's Republic of China
| | - Yuying Wu
- Department of General Practice, The Affiliated Luohu Hospital of Shenzhen University Medical School, Shenzhen, Guangdong, People's Republic of China; Department of Biostatistics and Epidemiology, School of Public Health, Shenzhen University Medical School, Shenzhen, Guangdong, People's Republic of China
| | - Xi Li
- Department of Epidemiology and Health Statistics, College of Public Health, Zhengzhou University, Zhengzhou, Henan, People's Republic of China
| | - Hao Huang
- Department of General Practice, The Affiliated Luohu Hospital of Shenzhen University Medical School, Shenzhen, Guangdong, People's Republic of China
| | - Fulan Hu
- Department of General Practice, The Affiliated Luohu Hospital of Shenzhen University Medical School, Shenzhen, Guangdong, People's Republic of China; Department of Biostatistics and Epidemiology, School of Public Health, Shenzhen University Medical School, Shenzhen, Guangdong, People's Republic of China
| | - Dongsheng Hu
- Department of General Practice, The Affiliated Luohu Hospital of Shenzhen University Medical School, Shenzhen, Guangdong, People's Republic of China; Department of Biostatistics and Epidemiology, School of Public Health, Shenzhen University Medical School, Shenzhen, Guangdong, People's Republic of China; Department of Epidemiology and Health Statistics, College of Public Health, Zhengzhou University, Zhengzhou, Henan, People's Republic of China
| | - Ming Zhang
- Department of General Practice, The Affiliated Luohu Hospital of Shenzhen University Medical School, Shenzhen, Guangdong, People's Republic of China.
| |
Collapse
|
6
|
Rehues P, Girona J, Guardiola M, Plana N, Scicali R, Piro S, Muñiz-Grijalvo O, Díaz-Díaz JL, Recasens L, Pinyol M, Rosales R, Esteban Y, Amigó N, Masana L, Ibarretxe D, Ribalta J. PCSK9 Inhibitors Have Apolipoprotein C-III-Related Anti-Inflammatory Activity, Assessed by 1H-NMR Glycoprotein Profile in Subjects at High or very High Cardiovascular Risk. Int J Mol Sci 2023; 24:2319. [PMID: 36768645 PMCID: PMC9917120 DOI: 10.3390/ijms24032319] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Revised: 01/12/2023] [Accepted: 01/16/2023] [Indexed: 01/26/2023] Open
Abstract
Atherosclerosis is a chronic inflammatory disease caused by the accumulation of cholesterol in the intima. Proprotein convertase subtilisin/kexin type 9 inhibitors (iPCSK9) can reduce low-density lipoprotein (LDL) cholesterol levels by 60%, but there is still no evidence that they can lower markers of systemic inflammation such as high-sensitivity C-reactive protein (hsCRP). Acute-phase serum glycoproteins are upregulated in the liver during systemic inflammation, and their role as inflammatory biomarkers is under clinical evaluation. In this observational study, we evaluate the effects of iPCSK9 on glycoproteins (Glyc) A, B and F. Thirty-nine patients eligible for iPCSK9 therapy were enrolled. One sample before and after one to six months of iPCSK9 therapy with alirocumab was obtained from each patient. Lipids, apolipoproteins, hsCRP and PCSK9 levels were measured by biochemical analyses, and the lipoprotein and glycoprotein profiles were measured by 1H nuclear magnetic resonance (1H-NMR). The PCSK9 inhibitor reduced total (36.27%, p < 0.001), LDL (55.05%, p < 0.001) and non-high-density lipoprotein (HDL) (45.11%, p < 0.001) cholesterol, apolipoprotein (apo) C-III (10%, p < 0.001), triglycerides (9.92%, p < 0.001) and glycoprotein signals GlycA (11.97%, p < 0.001), GlycB (3.83%, p = 0.017) and GlycF (7.26%, p < 0.001). It also increased apoA-I (2.05%, p = 0.043) and HDL cholesterol levels (11.58%, p < 0.001). Circulating PCSK9 levels increased six-fold (626.28%, p < 0.001). The decrease in Glyc signals positively correlated with the decrease in triglycerides and apoC-III. In conclusion, in addition to LDL cholesterol, iPCSK9 therapy also induces a reduction in systemic inflammation measured by 1H-NMR glycoprotein signals, which correlates with a decrease in triglycerides and apoC-III.
Collapse
Affiliation(s)
- Pere Rehues
- Universitat Rovira i Virgili, Departament de Medicina i Cirurgia, Unitat de Recerca en Lípids i Arteriosclerosi, 43201 Reus, Spain
- Institut d’Investigació Sanitària Pere Virgili, 43204 Reus, Spain
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas, 28029 Madrid, Spain
| | - Josefa Girona
- Universitat Rovira i Virgili, Departament de Medicina i Cirurgia, Unitat de Recerca en Lípids i Arteriosclerosi, 43201 Reus, Spain
- Institut d’Investigació Sanitària Pere Virgili, 43204 Reus, Spain
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas, 28029 Madrid, Spain
| | - Montse Guardiola
- Universitat Rovira i Virgili, Departament de Medicina i Cirurgia, Unitat de Recerca en Lípids i Arteriosclerosi, 43201 Reus, Spain
- Institut d’Investigació Sanitària Pere Virgili, 43204 Reus, Spain
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas, 28029 Madrid, Spain
| | - Núria Plana
- Institut d’Investigació Sanitària Pere Virgili, 43204 Reus, Spain
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas, 28029 Madrid, Spain
- Unitat de Medicina Vascular i Metabolisme, Servei de Medicina Interna, Hospital Universitari Sant Joan de Reus, 43204 Reus, Spain
| | - Roberto Scicali
- Department of Clinical and Experimental Medicine, University of Catania, 95131 Catania, Italy
| | - Salvatore Piro
- Department of Clinical and Experimental Medicine, University of Catania, 95131 Catania, Italy
| | - Ovidio Muñiz-Grijalvo
- Unidad Clinico-Experimental de Riesgo Vascular, Hospital Virgen del Rocío, 41013 Sevilla, Spain
| | - José Luis Díaz-Díaz
- Department of Internal Medicine, Complejo Hospitalario Universitario A Coruña, 15006 A Coruña, Spain
| | - Lluís Recasens
- Heart Diseases Biomedical Research Group, IMIM (Hospital del Mar Medical Research Institute), 08003 Barcelona, Spain
- Cardiac Rehabilitation Unit, Department of Cardiology, Hospital del Mar, 08003 Barcelona, Spain
| | - Marta Pinyol
- Universitat Rovira i Virgili, Departament de Medicina i Cirurgia, Unitat de Recerca en Lípids i Arteriosclerosi, 43201 Reus, Spain
| | - Roser Rosales
- Universitat Rovira i Virgili, Departament de Medicina i Cirurgia, Unitat de Recerca en Lípids i Arteriosclerosi, 43201 Reus, Spain
- Institut d’Investigació Sanitària Pere Virgili, 43204 Reus, Spain
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas, 28029 Madrid, Spain
| | - Yaiza Esteban
- Universitat Rovira i Virgili, Departament de Medicina i Cirurgia, Unitat de Recerca en Lípids i Arteriosclerosi, 43201 Reus, Spain
- Institut d’Investigació Sanitària Pere Virgili, 43204 Reus, Spain
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas, 28029 Madrid, Spain
| | - Núria Amigó
- Institut d’Investigació Sanitària Pere Virgili, 43204 Reus, Spain
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas, 28029 Madrid, Spain
- Biosfer Teslab, 43201 Reus, Spain
| | - Lluís Masana
- Universitat Rovira i Virgili, Departament de Medicina i Cirurgia, Unitat de Recerca en Lípids i Arteriosclerosi, 43201 Reus, Spain
- Institut d’Investigació Sanitària Pere Virgili, 43204 Reus, Spain
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas, 28029 Madrid, Spain
- Unitat de Medicina Vascular i Metabolisme, Servei de Medicina Interna, Hospital Universitari Sant Joan de Reus, 43204 Reus, Spain
| | - Daiana Ibarretxe
- Universitat Rovira i Virgili, Departament de Medicina i Cirurgia, Unitat de Recerca en Lípids i Arteriosclerosi, 43201 Reus, Spain
- Institut d’Investigació Sanitària Pere Virgili, 43204 Reus, Spain
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas, 28029 Madrid, Spain
- Unitat de Medicina Vascular i Metabolisme, Servei de Medicina Interna, Hospital Universitari Sant Joan de Reus, 43204 Reus, Spain
| | - Josep Ribalta
- Universitat Rovira i Virgili, Departament de Medicina i Cirurgia, Unitat de Recerca en Lípids i Arteriosclerosi, 43201 Reus, Spain
- Institut d’Investigació Sanitària Pere Virgili, 43204 Reus, Spain
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas, 28029 Madrid, Spain
| |
Collapse
|
7
|
Khalifeh M, Santos RD, Oskuee RK, Badiee A, Aghaee-Bakhtiari SH, Sahebkar A. A novel regulatory facet for hypertriglyceridemia: The role of microRNAs in the regulation of triglyceride-rich lipoprotein biosynthesis. Prog Lipid Res 2023; 89:101197. [PMID: 36400247 DOI: 10.1016/j.plipres.2022.101197] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2022] [Revised: 11/04/2022] [Accepted: 11/08/2022] [Indexed: 11/17/2022]
Abstract
Atherosclerotic cardiovascular disease (ASCVD) is one of the major leading global causes of death. Genetic and epidemiological studies strongly support the causal association between triacylglycerol-rich lipoproteins (TAGRL) and atherogenesis, even in statin-treated patients. Recent genetic evidence has clarified that variants in several key genes implicated in TAGRL metabolism are strongly linked to the increased ASCVD risk. There are several triacylglycerol-lowering agents; however, new therapeutic options are in development, among which are miRNA-based therapeutic approaches. MicroRNAs (miRNAs) are small non-coding RNAs (18-25 nucleotides) that negatively modulate gene expression through translational repression or degradation of target mRNAs, thereby reducing the levels of functional genes. MiRNAs play a crucial role in the development of hypertriglyceridemia as several miRNAs are dysregulated in both synthesis and clearance of TAGRL particles. MiRNA-based therapies in ASCVD have not yet been applied in human trials but are attractive. This review provides a concise overview of current interventions for hypertriglyceridemia and the development of novel miRNA and siRNA-based drugs. We summarize the miRNAs involved in the regulation of key genes in the TAGRLs synthesis pathway, which has gained attention as a novel target for therapeutic applications in CVD.
Collapse
Affiliation(s)
- Masoumeh Khalifeh
- Department of Medical Biotechnology and Nanotechnology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Raul D Santos
- Lipid Clinic Heart Institute (Incor), University of São Paulo, Medical School Hospital, São Paulo, Brazil
| | - Reza Kazemi Oskuee
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Ali Badiee
- Nanotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | | | - Amirhossein Sahebkar
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran; Biotechnology Research Centre, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; School of Medicine, The University of Western Australia, Perth, Australia; School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
8
|
Momtazi-Borojeni AA, Banach M, Ruscica M, Sahebkar A. The role of PCSK9 in NAFLD/NASH and therapeutic implications of PCSK9 inhibition. Expert Rev Clin Pharmacol 2022; 15:1199-1208. [PMID: 36193738 DOI: 10.1080/17512433.2022.2132229] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
INTRODUCTION There are inconsistent findings regarding the effect of lipid-lowering agents on nonalcoholic fatty liver disease (NAFLD). Proprotein Convertase Subtilisin/Kexin type 9 (PCSK9) is an important player in cholesterol homeostasis and intracellular lipogenesis, and PCSK9 inhibitors (PCSK9-i) have been found to be efficient for pharmacological management of hyperlipidemia. AREAS COVERED Whether PCSK9 (itself) or PCSK9-i affects NAFLD is still disputed. To address this question, we review published preclinical and clinical studies providing evidence for the role of PCSK9 in and the effect of PCSK9-I on the development and pathogenesis of NAFLD. EXPERT OPINION The current evidence from a landscape of preclinical and clinical studies examining the role of PCSK9 in NAFLD shows controversial results. Preclinical studies indicate that PCSK9 associates with NAFLD and nonalcoholic steatohepatitis (NASH) progression in opposite directions. In humans, it has been concluded that the severity of hepatic steatosis affects the correlation between circulating PCSK9 and liver fat content in humans, with a possible impact of circulating PCSK9 in the early stages of NAFLD, but not in the late stages. However, data from clinical trials with PCSK9-i reassure to the safety of these agents, although real-life long-term evidence is needed.
Collapse
Affiliation(s)
| | - Maciej Banach
- Department of Hypertension, Medical University of Lodz (MUL), Lodz, Poland.,Cardiovascular Research Centre, University of Zielona Gora, Zielona Gora, Poland
| | - Massimiliano Ruscica
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milan, Italy
| | - Amirhossein Sahebkar
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran.,Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.,School of Medicine, The University of Western Australia, Perth, Australia.,Department of Biotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
9
|
Sarkar SK, Matyas A, Asikhia I, Hu Z, Golder M, Beehler K, Kosenko T, Lagace TA. Pathogenic gain-of-function mutations in the prodomain and C-terminal domain of PCSK9 inhibit LDL binding. Front Physiol 2022; 13:960272. [PMID: 36187800 PMCID: PMC9515655 DOI: 10.3389/fphys.2022.960272] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Accepted: 08/23/2022] [Indexed: 11/30/2022] Open
Abstract
Proprotein convertase subtilisin/kexin type-9 (PCSK9) is a secreted protein that binds and mediates endo-lysosomal degradation of low-density lipoprotein receptor (LDLR), limiting plasma clearance of cholesterol-rich LDL particles in liver. Gain-of-function (GOF) point mutations in PCSK9 are associated with familial hypercholesterolemia (FH). Approximately 30%–40% of PCSK9 in normolipidemic human plasma is bound to LDL particles. We previously reported that an R496W GOF mutation in a region of PCSK9 known as cysteine-histidine–rich domain module 1 (CM1) prevents LDL binding in vitro [Sarkar et al., J. Biol. Chem. 295 (8), 2285–2298 (2020)]. Herein, we identify additional GOF mutations that inhibit LDL association, localized either within CM1 or a surface-exposed region in the PCSK9 prodomain. Notably, LDL binding was nearly abolished by a prodomain S127R GOF mutation, one of the first PCSK9 mutations identified in FH patients. PCSK9 containing alanine or proline substitutions at amino acid position 127 were also defective for LDL binding. LDL inhibited cell surface LDLR binding and degradation induced by exogenous PCSK9-D374Y but had no effect on an S127R-D374Y double mutant form of PCSK9. These studies reveal that multiple FH-associated GOF mutations in two distinct regions of PCSK9 inhibit LDL binding, and that the Ser-127 residue in PCSK9 plays a critical role.
Collapse
Affiliation(s)
- Samantha K. Sarkar
- Department of Biochemistry, Microbiology, and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
- University of Ottawa Heart Institute, Ottawa, ON, Canada
| | - Angela Matyas
- Department of Biochemistry, Microbiology, and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
- University of Ottawa Heart Institute, Ottawa, ON, Canada
| | - Ikhuosho Asikhia
- Department of Biochemistry, Microbiology, and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
- University of Ottawa Heart Institute, Ottawa, ON, Canada
| | - Zhenkun Hu
- University of Ottawa Heart Institute, Ottawa, ON, Canada
| | - Mia Golder
- University of Ottawa Heart Institute, Ottawa, ON, Canada
| | | | - Tanja Kosenko
- University of Ottawa Heart Institute, Ottawa, ON, Canada
| | - Thomas A. Lagace
- Department of Biochemistry, Microbiology, and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
- University of Ottawa Heart Institute, Ottawa, ON, Canada
- *Correspondence: Thomas A. Lagace,
| |
Collapse
|
10
|
Dafnis I, Tsouka AN, Gkolfinopoulou C, Tellis CC, Chroni A, Tselepis AD. PCSK9 is minimally associated with HDL but impairs the anti-atherosclerotic HDL effects on endothelial cell activation. J Lipid Res 2022; 63:100272. [PMID: 36067830 PMCID: PMC9526147 DOI: 10.1016/j.jlr.2022.100272] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Revised: 07/30/2022] [Accepted: 07/30/2022] [Indexed: 11/30/2022] Open
Abstract
Proprotein Convertase Subtilisin/Kexin type 9 (PCSK9) regulates the cell-surface localization of LDL receptors in hepatocytes and is associated with LDL and lipoprotein(a) [Lp(a)] uptake, reducing blood concentrations. However, the connection between PCSK9 and HDL is unclear. Here, we investigated the association of plasma PCSK9 with HDL subpopulations and examined the effects of PCSK9 on the atheroprotective function of HDL. We examined the association of PCSK9 with HDL in apoB-depleted plasma by ELISA, native PAGE, and immunoblotting. Our analyses showed that upon apoB-depletion, total circulating PCSK9 levels were 32% of those observed in normolipidemic plasma, and only 6% of PCSK9 in the apoB-depleted plasma, including both the mature and furin-cleaved forms, was associated with HDL. We also show human recombinant PCSK9 abolished the capacity of reconstituted HDL to reduce the formation of ROS in endothelial cells, while a PCSK9-blocking antibody enhanced the capacity of human HDL (in apoB-depleted plasma) to reduce ROS formation in endothelial cells and promote endothelial cell migration. Overall, our findings suggest that PCSK9 is only minimally associated with HDL particles, but PCSK9 in apoB-depleted plasma can affect the atheroprotective properties of HDL related to preservation of endothelial function. This study contributes to the elucidation of the pathophysiological role of plasma PCSK9 and highlights further the anti-atherosclerotic effect of PCSK9 inhibition.
Collapse
Affiliation(s)
- Ioannis Dafnis
- Institute of Biosciences and Applications, National Center for Scientific Research "Demokritos", Agia Paraskevi, Athens, Greece
| | - Aikaterini N Tsouka
- Atherothrombosis Research Centre, Department of Chemistry, University of Ioannina, Ioannina, Greece
| | - Christina Gkolfinopoulou
- Institute of Biosciences and Applications, National Center for Scientific Research "Demokritos", Agia Paraskevi, Athens, Greece
| | - Constantinos C Tellis
- Atherothrombosis Research Centre, Department of Chemistry, University of Ioannina, Ioannina, Greece
| | - Angeliki Chroni
- Institute of Biosciences and Applications, National Center for Scientific Research "Demokritos", Agia Paraskevi, Athens, Greece
| | - Alexandros D Tselepis
- Atherothrombosis Research Centre, Department of Chemistry, University of Ioannina, Ioannina, Greece.
| |
Collapse
|
11
|
Sadik NA, Rashed LA, El-Sawy SS. The Relationship of Circulating Proprotein Convertase Subtilisin/Kexin Type 9 With TSH and Lipid Profile in Newly Diagnosed Patients With Subclinical and Overt Hypothyroidism. Clin Med Insights Endocrinol Diabetes 2022; 15:11795514221093317. [PMID: 35494422 PMCID: PMC9039449 DOI: 10.1177/11795514221093317] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2021] [Accepted: 03/11/2022] [Indexed: 11/25/2022] Open
Abstract
Introduction: Overt and subclinical hypothyroidism are mostly associated with dyslipidemia, an essential cardiovascular risk factor. Recently, thyroid stimulating hormone (TSH) was identified to have a direct role on lipid metabolism via increased expression of hepatic proprotein convertase subtilisin/kexin type 9 (PCSK9). PCSK9 plays a crucial role in lipid metabolism via regulating LDL-C levels. Thus, we aimed to evaluate circulating PCSK9 levels and to assess its relationship with serum TSH and lipids in newly diagnosed patients had overt and subclinical hypothyroidism. Methods: In our study, we enrolled 60 newly diagnosed untreated patients with overt and subclinical hypothyroidism and 30 euthyroid subjects served as the control group. Serum TSH, FT4, FT3, lipid profile and circulating PCSK9 levels using ELISA kits were measured in all subjects. Our data were summarized using mean ± SD or median and interquartile range. Correlations between PCSK9 expression levels and different variables were done using Spearman correlation coefficient. Results: Circulating PCSK9 median levels were significantly increased in patients had overt and subclinical hypothyroidism (12.45 ng/ml, 7.50 ng/ml respectively) compared to the control group (3.30 ng/ml) (P < .001). Circulating PCSK9 levels significantly correlated positively with TSH, total cholesterol, triglycerides, and BMI, and negatively correlated with FT4 and FT3 among all studied subjects. Using multivariate regression analysis TSH was the only significant independent predictor of circulating PCSK9 (P < .001). Conclusion: Our results supports the new implication of TSH in lipid metabolism via the significant association with PCSK9. Whether this relationship between TSH and PCSK9 is a cause or just an association needs further evaluation.
Collapse
Affiliation(s)
- Noha Adly Sadik
- Internal Medicine Department, Diabetes and Endocrinology division, Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Laila Ahmed Rashed
- Biochemistry and Molecular Biology Department, Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Shereen Sadik El-Sawy
- Internal Medicine Department, Diabetes and Endocrinology division, Faculty of Medicine, Cairo University, Cairo, Egypt
| |
Collapse
|
12
|
Wang Z, Chen X, Liu J, Wang Y, Zhang S. Inclisiran inhibits oxidized low-density lipoprotein-induced foam cell formation in Raw264.7 macrophages via activating the PPARγ pathway. Autoimmunity 2022; 55:223-232. [PMID: 35289693 DOI: 10.1080/08916934.2022.2051142] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
Proprotein convertase subtilisin kexin type 9 (PCSK9) is a well-known proprotein convertase that influences foam cell formation and modulates atherosclerosis. Inclisiran is a novel chemosynthetic small interfering RNA that inhibits PCSK9 synthesis. This study aimed to explore the effect of inclisiran on oxidized low-density lipoprotein (ox-LDL)-induced foam cell formation in Raw264.7 macrophages and to investigate the underlying mechanisms. Raw264.7 cells were treated with ox-LDL to induce the formation of macrophage-derived foam cells. Oil Red O staining and high-performance liquid chromatography were performed to detect lipid accumulation and cholesterol levels. Dil-ox-LDL uptake assay, CCK-8, RT-qPCR, and Western blotting analysis were performed to examine ox-LDL uptake, cell viability, and expression of scavenger receptor-related factors. Inclisiran reduced lipid accumulation in ox-LDL-treated macrophages in a dose-dependent manner. Inclisiran significantly inhibited the levels of total cholesterol, free cholesterol, and cholesterol ester in the supernatant of Raw264.7 cells. Inclisiran reduced ox-LDL uptake and increased Raw264.7 cell viability. Meanwhile, inclisiran downregulated the expression of SR-A, LOX-1, and CD36 and upregulated SR-BI, ApoE, and ABCA1. Furthermore, inclisiran increased PPARγ activity and decreased NF-κB activity. An inhibitor of PPARγ (T0070907) reversed the beneficial effects of inclisiran on ox-LDL uptake, NF-κB inactivation, and cytokine expression. In conclusion, these data suggested that inclisiran inhibited the formation of macrophage-derived foam cells by activating the PPARγ pathway.HighlightsInclisiran reduces lipid accumulation in Raw264.7 cells;Inclisiran reduces ox-LDL uptake and increases Raw264.7 cell viability;Inclisiran inhibits foam cell formation by activating the PPARγ pathway.
Collapse
Affiliation(s)
- Zhaoping Wang
- Department of Emergency, Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University, Qingdao, P.R. China
| | - Xiangyu Chen
- Department of Emergency, Weihai Municipal Hospital, Weihai, P.R. China
| | - Jingxing Liu
- Emergency Department, Qingdao Municipal Hospital (Group), Qingdao NO.9 People's Hospital, Qingdao, P.R. China
| | - Yingcui Wang
- Department of Cardiology, Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University, Qingdao, P.R. China
| | - Suhua Zhang
- Department of Geriatrics, Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University, Qingdao, P.R. China
| |
Collapse
|
13
|
Triglyceride-rich lipoprotein and LDL particle subfractions and their association with incident type 2 diabetes: the PREVEND study. Cardiovasc Diabetol 2021; 20:156. [PMID: 34321006 PMCID: PMC8320057 DOI: 10.1186/s12933-021-01348-w] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Accepted: 07/20/2021] [Indexed: 12/29/2022] Open
Abstract
Background Triglyceride-rich lipoproteins particles (TRLP) and low density lipoprotein particles (LDLP) vary in size. Their association with β-cell function is not well described. We determined associations of TRLP and LDLP subfractions with β-cell function, estimated as HOMA-β, and evaluated their associations with incident T2D in the general population. Methods We included 4818 subjects of the Prevention of Renal and Vascular End-Stage Disease (PREVEND) study without T2D at baseline. TRLP and LDLP subfraction concentrations and their average sizes were measured using the LP4 algorithm of the Vantera nuclear magnetic resonance platform. HOMA-IR was used as measure of insulin resistance. HOMA-β was used as a proxy of β-cell function. Results In subjects without T2D at baseline, very large TRLP, and LDL size were inversely associated with HOMA-β, whereas large TRLP were positively associated with HOMA-β when taking account of HOMA-IR. During a median follow-up of 7.3 years, 263 participants developed T2D. In multivariable-adjusted Cox regression models, higher concentrations of total, very large, large, and very small TRLP (reflecting remnants lipoproteins) and greater TRL size were associated with an increased T2D risk after adjustment for relevant covariates, including age, sex, BMI, HDL-C, HOMA-β, and HOMA-IR. On the contrary, higher concentrations of large LDLP and greater LDL size were associated with a lower risk of developing T2D. Conclusions Specific TRL and LDL particle characteristics are associated with β-cell function taking account of HOMA-IR. Moreover, TRL and LDL particle characteristics are differently associated with incident T2D, even when taking account of HOMA-β and HOMA-IR. Supplementary Information The online version contains supplementary material available at 10.1186/s12933-021-01348-w.
Collapse
|
14
|
Farmaki P, Damaskos C, Garmpis N, Garmpi A, Savvanis S, Diamantis E. PCSK9 Inhibitors and Cardiovascular Disease: Impact on Cardiovascular Outcomes. Curr Drug Discov Technol 2021; 17:138-146. [PMID: 30526464 DOI: 10.2174/1570163816666181211112358] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2018] [Revised: 10/21/2018] [Accepted: 11/09/2018] [Indexed: 12/20/2022]
Abstract
Cardiovascular Disease (CVD) remains the leading cause of morbidity and mortality in the western world. Hypolipidemic drugs have long been used for the primary and secondary prevention of heart disease. However, the high frequency of recurrent events in patients despite hypolipidemic therapy has increased the need for new more targeted therapeutic approaches. Proprotein convertase subtilisin/kexin type 9 (PCSK9) inhibitors are monoclonal antibodies to the PCSK9 gene and represent a new class of drugs that have been shown to further decrease LDL-C when administered as a monotherapy or in combination with statins. In addition to LDL reduction, PCSK9 inhibitors are shown to decrease apolipoprotein B and lipoprotein (a) levels without major adverse effects. Whether or not PCSK9 inhibitors can actually reduce the incidence of cardiovascular events and ameliorate CVD prognosis is yet to be clarified. This review summarizes recent literature on the safety and efficacy of PCSK9 inhibitors on CVD outcome and its potential role in the management of patients with high-risk cardiovascular disease.
Collapse
Affiliation(s)
- Paraskevi Farmaki
- First Department of Pediatrics, Aghia Sophia Children's Hospital, Athens, Greece
| | - Christos Damaskos
- Second Department of Propedeutic Surgery, Laiko General Hospital, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Nikolaos Garmpis
- Second Department of Propedeutic Surgery, Laiko General Hospital, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Anna Garmpi
- Internal Medicine Department, Laiko General Hospital, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Spyridon Savvanis
- Department of Internal Medicine, General Hospital of Athens "Elpis", Athens, Greece
| | - Evangelos Diamantis
- Department of Endocrinology, Diabetes and Metabolism, Evangelismos Hospital, Athens, Greece
| |
Collapse
|
15
|
Shi J, Li X, Zhang W, Niu Y, Lin N, Zhang H, Ning G, Fan J, Qin L, Su Q, Yang Z. Circulating Proprotein Convertase Subtilisin/Kexin Type 9 Levels and Cardiometabolic Risk Factors: A Population-Based Cohort Study. Front Cardiovasc Med 2021; 8:664583. [PMID: 34041285 PMCID: PMC8141620 DOI: 10.3389/fcvm.2021.664583] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Accepted: 04/09/2021] [Indexed: 11/16/2022] Open
Abstract
Aims: To evaluate the prospective association of circulating PCSK9 levels with the cardiometabolic risk profiles (high LDL-cholesterol, high triglycerides, low HDL-cholesterol, hypertension, type 2 diabetes, and metabolic syndrome). Methods: A population-based prospective study was conducted among 7,104 Chinese individuals (age 56.2 ± 7.5 years; 32.0% men). Circulating PCSK9 levels were measured using ELISA. Results: Circulating PCSK9 levels were higher in women than men (286.7 ± 90.1 vs. 276.1 ± 86.4 ng/ml, p < 0.001). And circulating PCSK9 was positively correlated with LDL-cholesterol, total cholesterol, and triglycerides both in men and women (all p < 0.001). The positive correlation between PCSK9 and waist circumference, fasting glucose, insulin resistance, systolic blood pressure, diastolic blood pressure and C-reactive protein (all p < 0.01) was observed in women only. According to Cox regression analysis, circulating PCSK9 was positively associated with incidence of high LDL-cholesterol both in men (HR 1.33, 95% CI 1.09-1.65, p < 0.001) and women (HR 1.36, 95% CI 1.12-1.69, p < 0.001). Moreover, PCSK9 was significantly associated with incident high triglycerides (HR 1.31, 95% CI 1.13-1.72, p < 0.001), hypertension (HR 1.28, 95% CI 1.08-1.53, p = 0.011), type 2 diabetes (HR 1.34, 95% CI 1.09-1.76, p = 0.005), and metabolic syndrome (HR 1.30, 95% CI 1.11-1.65, p = 0.009) per SD change in women only. No statistically significant association was observed between circulating PCSK9 and incidence of low HDL-cholesterol (p > 0.1). Conclusions: Elevated circulating PCSK9 was significantly associated with cardiometabolic risk factors and independently contributed to the prediction of cardiometabolic risks in women.
Collapse
Affiliation(s)
- Jie Shi
- Department of Endocrinology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiaoyong Li
- Department of Endocrinology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Weiwei Zhang
- Department of Endocrinology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yixin Niu
- Department of Endocrinology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ning Lin
- Department of Endocrinology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hongmei Zhang
- Department of Endocrinology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Guang Ning
- Department of Endocrine and Metabolic Diseases, Shanghai Clinical Center for Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Institute of Endocrinology and Metabolism, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jiangao Fan
- Shanghai Key Laboratory of Children's Digestion and Nutrition, Department of Gastroenterology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Li Qin
- Department of Endocrinology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Qing Su
- Department of Endocrinology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhen Yang
- Department of Endocrinology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
16
|
PCSK9 Levels and Metabolic Profiles in Elderly Subjects with Different Glucose Tolerance under Statin Therapy. J Clin Med 2021; 10:jcm10050994. [PMID: 33801208 PMCID: PMC7957894 DOI: 10.3390/jcm10050994] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Revised: 02/15/2021] [Accepted: 02/22/2021] [Indexed: 12/15/2022] Open
Abstract
Proprotein convertase subtilisin/kexin type 9 (PCSK9) degrades low-density lipoprotein cholesterol (LDL-C) receptors, and thus regulates the LDL-C levels in the circulation. Type 2 diabetics often have elevated LDL-C levels. However, the functions of PCSK9 in patients with alterations of glu-cose metabolism and statin therapy are still unclear. Method: we investigated a large cohort of 608 subjects, born in 1945 in Oulu, Finland (Oulu Cohort 1945). We studied the effects of PSCK9 lev-els with different glucose tolerances (normal glucose tolerance (NGT), prediabetes (PreDM) or type 2 diabetes (T2D)) with and without statin medication, and analyzed clinical data, NMR metabolomics and PCSK9 plasma levels. Results: PCSK9 plasma levels did not significantly differ between the three groups. Statin therapy significantly increased the PCSK9 levels in NGT, PreDM and T2D groups compared with subjects with no statins. In the NGT group, negative associations between PCSK9 and LDL-C, intermediate-density lipoprotein cholesterol (IDL-C), very low-density lipoprotein cholesterol (VLDL-C), total cholesterol and LDL and IDL triglycerides were observed under statin medication. In contrast, in the PreDM and T2D groups, these associa-tions were lost. Conclusions: our data suggest that in subjects with abnormal glucose metabolism and statin therapy, the significant PCSK9-mediated effects on the lipid metabolites are lost com-pared to NGT subjects, but statins reduced the LDL-C and VLDL-C levels.
Collapse
|
17
|
Macchi C, Greco MF, Botta M, Sperandeo P, Dongiovanni P, Valenti L, Cicero AFG, Borghi C, Lupo MG, Romeo S, Corsini A, Magni P, Ferri N, Ruscica M. Leptin, Resistin, and Proprotein Convertase Subtilisin/Kexin Type 9: The Role of STAT3. THE AMERICAN JOURNAL OF PATHOLOGY 2020; 190:2226-2236. [PMID: 32798443 DOI: 10.1016/j.ajpath.2020.07.016] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/23/2020] [Revised: 07/17/2020] [Accepted: 07/30/2020] [Indexed: 01/13/2023]
Abstract
In a condition of dysfunctional visceral fat depots, as in the case of obesity, alterations in adipokine levels may be detrimental for the cardiovascular system. The proinflammatory leptin and resistin adipokines have been described as possible links between obesity and atherosclerosis. The present study was aimed at evaluating whether proprotein convertase subtilisin/kexin type 9 (PCSK9), a key regulator of low-density lipoprotein metabolism, is induced by leptin and resistin through the involvement of the inflammatory pathway of STAT3. In HepG2 cells, leptin and resistin up-regulated PCSK9 gene and protein expression, as well as the phosphorylation of STAT3. Upon STAT3 silencing, leptin and resistin lost their ability to activate PCSK9. The knockdown of STAT3 did not affect the expression of leptin and resistin receptors or that of PCSK9. The analysis of the human PCSK9 promoter region showed that the two adipokines raised PCSK9 promoter activity via the involvement of a sterol regulatory element motif. In healthy males, a positive association between circulating leptin and PCSK9 levels was found only when the body mass index was <25 kg/m2. In conclusion, this study identified STAT3 as one of the molecular regulators of leptin- and resistin-mediated transcriptional induction of PCSK9.
Collapse
Affiliation(s)
- Chiara Macchi
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milan, Italy.
| | - Maria Francesca Greco
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milan, Italy
| | - Margherita Botta
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milan, Italy
| | - Paola Sperandeo
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milan, Italy
| | - Paola Dongiovanni
- General Medicine and Metabolic Diseases, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Luca Valenti
- Department of Pathophysiology and Transplantation, Università degli Studi di Milano, Milan, Italy; Translational Medicine, Transfusion Medicine and Hematology, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Arrigo F G Cicero
- Department of Medicine and Surgery Sciences, Alma Mater Studiorum University of Bologna, Bologna, Italy
| | - Claudio Borghi
- Department of Medicine and Surgery Sciences, Alma Mater Studiorum University of Bologna, Bologna, Italy
| | - Maria Giovanna Lupo
- Department of Pharmaceutical and Pharmacological Sciences, Università degli Studi di Padova, Padova, Italy
| | - Stefano Romeo
- Department of Molecular and Clinical Medicine, University of Gothenburg, Gothenburg, Sweden; Clinical Nutrition Unit, Department of Medical and Surgical Science, Magna Graecia University, Catanzaro, Italy; Department of Cardiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Alberto Corsini
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milan, Italy; IRCCS MultiMedica, Sesto S. Giovanni, Milan, Italy
| | - Paolo Magni
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milan, Italy; IRCCS MultiMedica, Sesto S. Giovanni, Milan, Italy
| | - Nicola Ferri
- Department of Pharmaceutical and Pharmacological Sciences, Università degli Studi di Padova, Padova, Italy
| | - Massimiliano Ruscica
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milan, Italy.
| |
Collapse
|
18
|
Malvandi AM, Canclini L, Alliaj A, Magni P, Zambon A, Catapano AL. Progress and prospects of biological approaches targeting PCSK9 for cholesterol-lowering, from molecular mechanism to clinical efficacy. Expert Opin Biol Ther 2020; 20:1477-1489. [PMID: 32715821 DOI: 10.1080/14712598.2020.1801628] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
INTRODUCTION Cardiovascular disorders are one of the leading causes of mortality and morbidity worldwide. Recent advances showed a promising role of proprotein convertase subtilisin/kexin type 9 (PCSK9) as a critical player in regulating plasma LDL levels and lipid metabolism. AREAS COVERED This review addresses the molecular functions of PCSK9 with a vision on the clinical progress of utilizing monoclonal antibodies and other biological approaches to block PCSK9 activity. The successful clinical trials with monoclonal antibodies are reviewed. Recent advances in (pre)clinical trials of other biological approaches, such as small interfering RNAs, are also discussed. EXPERT OPINION Discovery of PCSK9 and clinical use of its inhibitors to manage lipid metabolism is a step forward in hypolipidaemic therapy. A better understanding of the molecular activity of PCSK9 can help to identify new approaches in the inhibition of PCSK9 expression/activity. Whether if PCSK9 plays a role in other cardiometabolic conditions may provide grounds for further development of therapies.
Collapse
Affiliation(s)
| | - Laura Canclini
- IRCCS Multimedica , Milan, Italy.,Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano , Milan, Italy
| | | | - Paolo Magni
- IRCCS Multimedica , Milan, Italy.,Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano , Milan, Italy
| | - Alberto Zambon
- IRCCS Multimedica , Milan, Italy.,Department of Medicine, Università degli Studi di Padova , Padua, Italy
| | - Alberico Luigi Catapano
- IRCCS Multimedica , Milan, Italy.,Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano , Milan, Italy
| |
Collapse
|
19
|
Paquette M, Gauthier D, Chamberland A, Prat A, De Lucia Rolfe E, Rasmussen JJ, Kaduka L, Seidah NG, Bernard S, Christensen DL, Baass A. Circulating PCSK9 is associated with liver biomarkers and hepatic steatosis. Clin Biochem 2020; 77:20-25. [PMID: 31972148 PMCID: PMC7614815 DOI: 10.1016/j.clinbiochem.2020.01.003] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Revised: 01/02/2020] [Accepted: 01/14/2020] [Indexed: 12/12/2022]
Abstract
BACKGROUND In parallel to the increasing prevalence of metabolic syndrome, the prevalence of hepatic steatosis has also increased dramatically worldwide. Hepatic steatosis is a major risk factor of hepatic cirrhosis, cardiovascular disease and type 2 diabetes. Circulating levels of proprotein convertase subtilisin/kexin type 9 (PCSK9) have been positively associated with the metabolic syndrome. However, the association between PCSK9 and the liver function is still controversial. OBJECTIVE The objective of this study is to investigate the association between circulating PCSK9 levels and the presence of hepatic steatosis, as well as with liver biomarkers in a cohort of healthy individuals. METHODS Total PCSK9 levels were measured by an in-house ELISA using a polyclonal antibody. Plasma albumin, alkaline phosphatase, ALT, AST, total bilirubin and GGT were measured in 698 individuals using the COBAS system. The presence of hepatic steatosis was assessed using ultrasound liver scans. RESULTS In a multiple regression model adjusted for age, sex, insulin resistance, body mass index and alcohol use, circulating PCSK9 level was positively associated with albumin (β = 0.102, P = 0.008), alkaline phosphatase (β = 0.201, P < 0.0001), ALT (β = 0.238, P < 0.0001), AST (β = 0.120, P = 0.003) and GGT (β = 0.103, P = 0.007) and negatively associated with total bilirubin (β = -0.150, P < 0.0001). Tertile of circulating PCSK9 was also associated with hepatic steatosis (OR 1.48, 95% CI 1.05-2.08, P = 0.02). CONCLUSION Our data suggest a strong association between PCSK9 and liver biomarkers as well as hepatic steatosis. Further studies are needed to explore the role of PCSK9 on hepatic function.
Collapse
Affiliation(s)
- Martine Paquette
- Lipids, Nutrition and Cardiovascular Prevention Clinic of the Montreal Clinical Research Institute, Montreal, Canada
| | - Dany Gauthier
- Lipids, Nutrition and Cardiovascular Prevention Clinic of the Montreal Clinical Research Institute, Montreal, Canada
| | - Ann Chamberland
- Laboratory of Biochemical Neuroendocrinology of the Montreal Clinical Research Institute, Montreal, Canada
| | - Annik Prat
- Laboratory of Biochemical Neuroendocrinology of the Montreal Clinical Research Institute, Montreal, Canada
| | - Emanuella De Lucia Rolfe
- Medical Research Council Epidemiology Unit, University of Cambridge School of Clinical Medicine, Institute of Metabolic Science, Cambridge Biomedical Campus, Cambridge, United Kingdom
| | - Jon J Rasmussen
- Centre of Endocrinology and Metabolism, Department of Internal Medicine, Copenhagen University Hospitals, Herlev and Gentofte, Denmark; Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen Copenhagen, Denmark
| | - Lydia Kaduka
- Centre for Public Health Research, KEMRI, Nairobi, Kenya
| | - Nabil G Seidah
- Laboratory of Biochemical Neuroendocrinology of the Montreal Clinical Research Institute, Montreal, Canada
| | - Sophie Bernard
- Lipids, Nutrition and Cardiovascular Prevention Clinic of the Montreal Clinical Research Institute, Montreal, Canada; Department of Medicine, Division of Endocrinology, Université de Montreal, Montreal, Canada
| | - Dirk L Christensen
- Medical Research Council Epidemiology Unit, University of Cambridge School of Clinical Medicine, Institute of Metabolic Science, Cambridge Biomedical Campus, Cambridge, United Kingdom; Department of Public Health, University of Copenhagen, Copenhagen, Denmark
| | - Alexis Baass
- Lipids, Nutrition and Cardiovascular Prevention Clinic of the Montreal Clinical Research Institute, Montreal, Canada; Department of Medicine, Divisions of Experimental Medicine and Medical Biochemistry, McGill University, Montreal, Canada.
| |
Collapse
|
20
|
Lee JS, Mukhopadhyay P, Matyas C, Trojnar E, Paloczi J, Yang YR, Blank BA, Savage C, Sorokin AV, Mehta NN, Vendruscolo JCM, Koob GF, Vendruscolo LF, Pacher P, Lohoff FW. PCSK9 inhibition as a novel therapeutic target for alcoholic liver disease. Sci Rep 2019; 9:17167. [PMID: 31748600 PMCID: PMC6868240 DOI: 10.1038/s41598-019-53603-6] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2019] [Accepted: 11/04/2019] [Indexed: 12/19/2022] Open
Abstract
Alcoholic liver disease (ALD) causes significant morbidity and mortality, and pharmacological treatment options are limited. In this study, we evaluated the PCSK9 inhibitor alirocumab, a monoclonal antibody that robustly reduces low-density lipoprotein cholesterol (LDL-C), for the treatment of ALD using a rat model of chronic alcohol exposure. Alirocumab (50 mg/kg) or vehicle was administered weekly for 6 weeks to rats receiving a 12% alcohol liquid diet or an isocaloric control diet. At the end of the alcohol exposure protocol, serum and liver samples were obtained for molecular characterization and histopathological analysis. PCSK9 inhibition with alirocumab attenuated alcohol-induced hepatic triglyceride accumulation through regulation of lipid metabolism (mRNA expression of modulators of fatty acid synthesis (FAS) and catabolism (PPARα and CPT1)), hepatocellular injury (ALT), hepatic inflammation (mRNA expression of pro-inflammatory cytokines/chemokines (TNFa, IL-1β, IL-22, IL-33, IL-17α, IL-2, MIP-2, and MCP-1), and neutrophil infiltration (myeloperoxidase staining)). Alirocumab treatment also attenuated alcohol-induced PCSK9 mRNA elevation and upregulated LDL-receptor (LDL-R) via modulation of the transcription factors (SREBP-1, SREBP-2, and E2F1) in liver. We demonstrated that chronic anti-PCSK9 treatment using the monoclonal antibody alirocumab attenuated alcohol-induced steatohepatitis in the rat model. Given the large unmet clinical need for effective and novel treatments for ALD, anti-PCSK9 treatment with the monoclonal antibody that spares liver metabolism is a viable new therapeutic possibility. Future studies are needed to elucidate the exact role of PCSK9 in ALD and alcohol use disorder (AUD) and to evaluate efficacy and safety of anti-PCSK9 treatment in clinical populations with ALD/AUD.
Collapse
Affiliation(s)
- Ji Soo Lee
- Section on Clinical Genomics and Experimental Therapeutics, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD, USA
| | - Partha Mukhopadhyay
- Laboratory of Cardiovascular Physiology and Tissue Injury, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD, USA
| | - Csaba Matyas
- Laboratory of Cardiovascular Physiology and Tissue Injury, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD, USA
| | - Eszter Trojnar
- Laboratory of Cardiovascular Physiology and Tissue Injury, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD, USA
| | - Janos Paloczi
- Laboratory of Cardiovascular Physiology and Tissue Injury, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD, USA
| | - Yuan Ru Yang
- Laboratory of Cardiovascular Physiology and Tissue Injury, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD, USA
| | - Brandon A Blank
- Neurobiology of Addiction Section, National Institute on Drug Abuse, National Institutes of Health, Baltimore, MD, USA
| | - Cody Savage
- Laboratory of Cardiovascular Physiology and Tissue Injury, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD, USA
| | - Alexander V Sorokin
- Lipoprotein Metabolism Section, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Nehal N Mehta
- Section of Inflammation and Cardiometabolic Diseases, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Janaina C M Vendruscolo
- Neurobiology of Addiction Section, National Institute on Drug Abuse, National Institutes of Health, Baltimore, MD, USA
| | - George F Koob
- Neurobiology of Addiction Section, National Institute on Drug Abuse, National Institutes of Health, Baltimore, MD, USA
| | - Leandro F Vendruscolo
- Neurobiology of Addiction Section, National Institute on Drug Abuse, National Institutes of Health, Baltimore, MD, USA
| | - Pal Pacher
- Laboratory of Cardiovascular Physiology and Tissue Injury, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD, USA
| | - Falk W Lohoff
- Section on Clinical Genomics and Experimental Therapeutics, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
21
|
Inclisiran-New hope in the management of lipid disorders? J Clin Lipidol 2019; 14:16-27. [PMID: 31879073 DOI: 10.1016/j.jacl.2019.11.001] [Citation(s) in RCA: 82] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2019] [Revised: 10/16/2019] [Accepted: 11/05/2019] [Indexed: 11/23/2022]
Abstract
Drugs reducing plasma concentrations of apolipoprotein B-containing lipoproteins have been demonstrated to reduce the risk of cardiovascular disease (CVD) in both primary and secondary prevention. Despite the demonstrated efficacy of statins and ezetimibe on low-density lipoprotein (LDL) concentration and long-term CVD risk, a large number of patients do not achieve their therapeutic goals. The introduction of monoclonal antibodies against proprotein convertase subtilisin/kexin type 9 (PCSK9) protein was a milestone in the treatment of lipid disorders, as their administration leads to unprecedentedly low LDL cholesterol concentrations. Inclisiran represents an entirely new mechanism of PSCK9 protein inhibition in hepatocytes, targeting the messenger RNA for PCSK9. Its administration is necessary only every 3 to 6 months, which is an essential advantage over statin and monoclonal antibody therapy. The infrequent administration regimen can increase the number of patients who maintain their therapeutic goals, especially in patients struggling to comply with daily or biweekly pharmacotherapy. Preclinical studies and Phase I and Phase II clinical trials of inclisiran have demonstrated its tolerability and efficacy in promoting long-term reduction of both PCSK9 protein and LDL cholesterol. The efficacy and safety of inclisiran will continue to be assessed in ongoing and forthcoming trials on larger patient groups. If the results of these trials reflect previously published data, they will add further evidence that inclisiran might be a revolutionary new tool in the pharmacologic management of plasma lipids. This review summarizes the currently available literature data on inclisiran with respect to its mechanism of action, effectiveness, and safety as a lipid-lowering drug for CVD prevention.
Collapse
|
22
|
Kolovou GD, Watts GF, Mikhailidis DP, Pérez-Martínez P, Mora S, Bilianou H, Panotopoulos G, Katsiki N, Ooi TC, Lopez-Miranda J, Tybjærg-Hansen A, Tentolouris N, Nordestgaard BG. Postprandial Hypertriglyceridaemia Revisited in the Era of Non-Fasting Lipid Profile Testing: A 2019 Expert Panel Statement, Narrative Review. Curr Vasc Pharmacol 2019; 17:515-537. [DOI: 10.2174/1570161117666190503123911] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2019] [Revised: 04/01/2019] [Accepted: 04/11/2019] [Indexed: 12/17/2022]
Abstract
Postprandial hypertriglyceridaemia, defined as an increase in plasma triglyceride-containing
lipoproteins following a fat meal, is a potential risk predictor of atherosclerotic cardiovascular disease
and other chronic diseases. Several non-modifiable factors (genetics, age, sex and menopausal status)
and lifestyle factors (diet, physical activity, smoking status, obesity, alcohol and medication use) may
influence postprandial hypertriglyceridaemia. This narrative review considers the studies published over
the last decade that evaluated postprandial hypertriglyceridaemia. Additionally, the genetic determinants
of postprandial plasma triglyceride levels, the types of meals for studying postprandial triglyceride response,
and underlying conditions (e.g. familial dyslipidaemias, diabetes mellitus, metabolic syndrome,
non-alcoholic fatty liver and chronic kidney disease) that are associated with postprandial hypertriglyceridaemia
are reviewed; therapeutic aspects are also considered.
Collapse
Affiliation(s)
- Genovefa D. Kolovou
- Cardiology Department and LDL-Apheresis Unit, Onassis Cardiac Surgery Center, Athens, Greece
| | - Gerald F. Watts
- Lipid Disorders Clinic, Department of Cardiology, Royal Perth Hospital, School of Medicine, Faculty of Health and Medical Sciences, University of Western Australia, Crawley, Australia
| | - Dimitri P. Mikhailidis
- Department of Clinical Biochemistry, Royal Free Hospital Campus, University College London Medical School, University College London (UCL), London, United Kingdom
| | - Pablo Pérez-Martínez
- Lipid and Atherosclerosis Unit, IMIBIC/Reina Sofia University Hospital/University of Cordoba, and CIBER Fisiopatologia Obesidad y Nutricion (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
| | - Samia Mora
- Center for Lipid Metabolomics, Divisions of Preventive and Cardiovascular Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, United States
| | - Helen Bilianou
- Department of Cardiology, Tzanio Hospital, Piraeus, Greece
| | | | - Niki Katsiki
- First Department of Internal Medicine, Division of Endocrinology-Metabolism, Diabetes Center, AHEPA University Hospital, Thessaloniki, Greece
| | - Teik C. Ooi
- Department of Medicine, Division of Endocrinology and Metabolism, University of Ottawa, Ottawa Hospital Research Institute, Ottawa, ON, Canada
| | - José Lopez-Miranda
- Lipid and Atherosclerosis Unit, IMIBIC/Reina Sofia University Hospital/University of Cordoba, and CIBER Fisiopatologia Obesidad y Nutricion (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
| | - Anne Tybjærg-Hansen
- Department of Clinical Biochemistry, Rigshospitalet, Copenhagen University Hospital, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Nicholas Tentolouris
- First Department of Propaedeutic Internal Medicine, Medical School, National and Kapodistrian University of Athens, Laiko General Hospital, Athens, Greece
| | - Børge G. Nordestgaard
- Department of Clinical Biochemistry, Herlev and Gentofte Hospital, Copenhagen University Hospital, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
23
|
Jeenduang N. Circulating PCSK9 concentrations are increased in postmenopausal women with the metabolic syndrome. Clin Chim Acta 2019; 494:151-156. [DOI: 10.1016/j.cca.2019.04.067] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2018] [Revised: 04/12/2019] [Accepted: 04/15/2019] [Indexed: 02/06/2023]
|
24
|
Case-control study on PCSK9 R496W (rs374603772) and D374Y (rs137852912) mutations in Turkish patients with primary dyslipidemia. Anatol J Cardiol 2019; 19:334-340. [PMID: 29724976 PMCID: PMC6280272 DOI: 10.14744/anatoljcardiol.2018.86648] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
Objective: The aim of this study was to investigate the relationships between F216L (rs28942112), R496W (rs374603772), S127R (rs28942111), and D374Y (rs137852912) PCSK9 gain-of-function (GOF) mutations and primary dyslipidemia and serum lipid levels in patients with primary dyslipidemia. Methods: In this case-control study, DNA was isolated from blood samples collected from patients diagnosed with primary dyslipidemia in cardiology outpatient clinic of Ege University (n=200) and healthy individuals (n=201). F216L, R496W, S127R, and D374Y GOF mutations in the PCSK9 gene were evaluated and genotyped according to the results of melting curve analysis performed in a real-time polymerase chain reaction (PCR) 480 instrument using specific primers for each mutation. Results: There were statistically significant differences between the patient and individuals in control groups in the R496W and D374Y mutations (χ2=10.742 p=0.005; χ2=6.078 p=0.048, respectively). In addition, triglyceride levels in patients with primary dyslipidemia heterozygous for R496W and D374Y mutations were 12.8-fold (p=0.015) and 3.4-fold (p=0.03) higher than that in mutant and wild-type genotype, respectively. Additionally, in the entire study group (n=401), PCSK9 R496W and D374Y mutation carriers had increased total cholesterol (p=0.021), triglycerides (p=0.0001), HDL cholesterol (p=0.028), and low-density lipoproteins (LDL) cholesterol (p=0.028) levels. However, F216L (rs28942112) and S127R (rs28942111) mutations were not detected in patients with primary dyslipidemia and healthy controls. Conclusion: We conclude that the PCSK9 R496W (rs374603772) and D374Y (rs137852912) GOF mutations may be significant risk factors in the development of primary dyslipidemia and may have significant impact on lipid parameters in general population.
Collapse
|
25
|
Delayed postprandial TAG peak after intake of SFA compared with PUFA in subjects with and without familial hypercholesterolaemia: a randomised controlled trial. Br J Nutr 2019; 119:1142-1150. [PMID: 29759104 DOI: 10.1017/s0007114518000673] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Postprandial hypertriacylglycerolaemia is associated with an increased risk of developing CVD. How fat quality influences postprandial lipid response is scarcely explored in subjects with familial hypercholesterolaemia (FH). The aim of this study was to investigate the postprandial response of TAG and lipid sub-classes after consumption of high-fat meals with different fat quality in subjects with FH compared with normolipidaemic controls. A randomised controlled double-blind cross-over study with two meals and two groups was performed. A total of thirteen hypercholesterolaemic subjects with FH who discontinued lipid-lowering treatment 4 weeks before and during the study, and fourteen normolipidaemic controls, were included. Subjects were aged 18-30 years and had a BMI of 18·5-30·0 kg/m2. Each meal consisted of a muffin containing 60 g (70 E%) of fat, either mainly SFA (40 E%) or PUFA (40 E%), eaten in a random order with a wash-out period of 3-5 weeks between the meals. Blood samples were collected at baseline (fasting) and 2, 4 and 6 h after intake of the meals. In both FH and control subjects, the level of TAG and the largest VLDL sub-classes peaked at 2 h after intake of PUFA and at 4 h after intake of SFA. No significant differences were found in TAG levels between meals or between groups (0·25≤P≤0·72). The distinct TAG peaks may reflect differences in the postprandial lipid metabolism after intake of fatty acids with different chain lengths and degrees of saturation. The clinical impact of these findings remains to be determined.
Collapse
|
26
|
Liu Y, Wang X, Han J, Liu L, Jin Y, Jin L, Ye P. PCSK9 positively correlates with plasma sdLDL in community-dwelling population but not in diabetic participants after confounder adjustment. Medicine (Baltimore) 2019; 98:e15062. [PMID: 30946354 PMCID: PMC6456037 DOI: 10.1097/md.0000000000015062] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
This study aimed to investigate the relationship between plasma proprotein convertase subtilisin kexin 9 (PCSK9) and small dense low-density lipoprptein (sdLDL) in diabetic and non-diabetic participants in a community-dwelling cohort.The plasma levels of PCSK9 and sdLDL were detected in 1766 participants (median age: 61.40 years; 733 males vs 1033 females; 383 diabetic vs 1383 non-diabetic patients) from the Pingguoyuan community of Beijing, China.Results showed that Pearson correlation analysis revealed a positive correlation between PCSK9 and sdLDL (r = 0.263, P < .001). Multiple linear regression analysis showed a significant positive correlation between plasma PCSK9 and sdLDL in the whole population study. sdLDL was used as the dependent variable, and the potential cofounders were adjusted. However, any independent relationship was not observed between circulating PCSK9 and sdLDL in the diabetic subpopulation (r = 0.269, P < .05, β = 9.591, P > .05).Thus, there is a positive correlation between plasma PCSK9 and sdLDL in a community-dwelling cohort, but not in type 2 diabetic subpopulation, after confounder adjustment.
Collapse
Affiliation(s)
- Yan Liu
- Department of Geriatric Cardiology, Chinese PLA General Hospital
- Critical Care Center, The 302 Hospital, People's Liberation Army
| | - Xiaona Wang
- Department of Geriatric Cardiology, Chinese PLA General Hospital
| | - Jie Han
- Department of Cardiology, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an
| | - Lu Liu
- Department of Geriatric Cardiology, Chinese PLA General Hospital
| | - Ying Jin
- Department of Geriatric Cardiology, Chinese PLA General Hospital
- Geriatric Institute, Chinese PLA Air Force General Hospital, Beijing, China
| | - Liyuan Jin
- Department of Geriatric Cardiology, Chinese PLA General Hospital
| | - Ping Ye
- Department of Geriatric Cardiology, Chinese PLA General Hospital
| |
Collapse
|
27
|
Shrestha P, van de Sluis B, Dullaart RP, van den Born J. Novel aspects of PCSK9 and lipoprotein receptors in renal disease-related dyslipidemia. Cell Signal 2019; 55:53-64. [DOI: 10.1016/j.cellsig.2018.12.001] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2018] [Revised: 12/01/2018] [Accepted: 12/03/2018] [Indexed: 12/12/2022]
|
28
|
Bojanin D, Vekic J, Milenkovic T, Vukovic R, Zeljkovic A, Stefanovic A, Janac J, Ivanisevic J, Mitrovic K, Miljkovic M, Spasojevic-Kalimanovska V. Association between proprotein convertase subtilisin/kexin 9 (PCSK9) and lipoprotein subclasses in children with type 1 diabetes mellitus: Effects of glycemic control. Atherosclerosis 2019; 280:14-20. [DOI: 10.1016/j.atherosclerosis.2018.11.020] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/11/2018] [Revised: 10/01/2018] [Accepted: 11/08/2018] [Indexed: 12/18/2022]
|
29
|
Rasmussen LD, Bøttcher M, Ivarsen P, Jørgensen HS, Nyegaard M, Buttenschøn H, Gustafsen C, Glerup S, Bøtker HE, Svensson M, Winther S. Association between circulating proprotein convertase subtilisin/kexin type 9 levels and prognosis in patients with severe chronic kidney disease. Nephrol Dial Transplant 2018; 35:632-639. [PMID: 30137516 DOI: 10.1093/ndt/gfy257] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2018] [Accepted: 07/05/2018] [Indexed: 01/18/2023] Open
Abstract
Abstract
Background
Chronic kidney disease is a risk factor for premature development of coronary atherosclerosis and mortality. A high level of proprotein convertase subtilisin/kexin type 9 (PCSK9) is a recently recognized cardiovascular risk factor and has become the target of effective inhibitory treatment. In 167 kidney transplantation candidates, we aimed to: (i) compare levels of PCSK9 with those of healthy controls, (ii) examine the association between levels of PCSK9 and low-density lipoprotein cholesterol (LDL-c) and the degree of coronary artery disease (CAD) and (iii) evaluate if levels of PCSK9 predict major adverse cardiac events (MACE) and mortality.
Methods
Kidney transplant candidates (n = 167) underwent coronary computed tomography angiography (CCTA) and invasive coronary angiography (ICA) before transplantation. MACE and mortality data were extracted from the Western Denmark Heart Registry, a review of patient records and patient interviews. A group of 79 healthy subjects were used as controls.
Results
Mean PCSK9 levels did not differ between healthy controls and kidney transplant candidates. In patients not receiving lipid-lowering therapy, PCSK9 correlated positively with LDL-c (rho = 0.24, P < 0.05). Mean PCSK9 was similar in patients with and without obstructive CAD at both CCTA and ICA. In a multiple regression analysis, PCSK9 was associated with neither LDL-c (β=−6.45, P = 0.44) nor coronary artery calcium score (β=2.17, P = 0.84). During a follow-up of 3.7 years, PCSK9 levels were not associated with either MACE or mortality.
Conclusions
The ability of PCSK9 levels to predict cardiovascular disease and prognosis does not seem to apply to a cohort of kidney transplant candidates.
Collapse
Affiliation(s)
| | - Morten Bøttcher
- Department of Cardiology, Regional Hospital Unit West Jutland, Herning, Denmark
| | - Per Ivarsen
- Department of Nephrology, Aarhus University Hospital, Aarhus, Denmark
| | | | - Mette Nyegaard
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Henriette Buttenschøn
- Department of Clinical Medicine, Aarhus University – Translational Neuropsychiatry Unit, Risskov, Denmark
| | | | - Simon Glerup
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Hans Erik Bøtker
- Department of Cardiology, Aarhus University Hospital, Aarhus, Denmark
| | - My Svensson
- Department of Nephrology, Akershus University Hospital, Lørenskog, Norway
- Institute of Clinical Medicine, University of Oslo, Søsterhjemmet, Oslo, Norway
| | - Simon Winther
- Department of Cardiology, Aarhus University Hospital, Aarhus, Denmark
| |
Collapse
|
30
|
Dijk W, Le May C, Cariou B. Beyond LDL: What Role for PCSK9 in Triglyceride-Rich Lipoprotein Metabolism? Trends Endocrinol Metab 2018; 29:420-434. [PMID: 29665987 DOI: 10.1016/j.tem.2018.03.013] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/05/2018] [Revised: 03/10/2018] [Accepted: 03/15/2018] [Indexed: 10/17/2022]
Abstract
Elevated plasma triglyceride (TG) levels are an independent risk factor for cardiovascular disease (CVD). Proprotein convertase subtilisin-kexin 9 (PCSK9) - a protein therapeutically targeted to lower plasma cholesterol levels - might regulate plasma TG-rich lipoprotein (TRL) levels. We provide a timely and critical review of the current evidence for a role of PCSK9 in TRL metabolism by assessing the impact of PCSK9 gene variants, by reviewing recent clinical data with PCSK9 inhibitors, and by describing the potential mechanisms by which PCSK9 might regulate TRL metabolism. We conclude that the impact of PCSK9 on TRL metabolism is relatively modest, especially compared to its impact on cholesterol metabolism.
Collapse
Affiliation(s)
- Wieneke Dijk
- L'institut du thorax, INSERM, CNRS, Université de Nantes, Nantes, France
| | - Cédric Le May
- L'institut du thorax, INSERM, CNRS, Université de Nantes, Nantes, France
| | - Bertrand Cariou
- L'institut du thorax, INSERM, CNRS, Université de Nantes, Nantes, France; L'institut du thorax, Department of Endocrinology, CHU NANTES, Nantes, France.
| |
Collapse
|
31
|
Kjellmo CA, Hovland A, Lappegård KT. CVD Risk Stratification in the PCSK9 Era: Is There a Role for LDL Subfractions? Diseases 2018; 6:diseases6020045. [PMID: 29861477 PMCID: PMC6023332 DOI: 10.3390/diseases6020045] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2018] [Revised: 05/23/2018] [Accepted: 05/24/2018] [Indexed: 12/22/2022] Open
Abstract
Proprotein convertase subtilisin/kexin type 9 (PCSK9) inhibitors reduce the risk of cardiovascular events and all-cause mortality in patients at high risk of cardiovascular disease (CVD). Due to high costs and unknown long-term adverse effects, critical evaluation of patients considered for PCSK9 inhibitors is important. It has been proposed that measuring low-density lipoprotein (LDL) subfractions, or LDL particle numbers (LDL-P), could be of value in CVD risk assessment and may identify patients at high risk of CVD. This review evaluates the evidence for the use of LDL subfractions, or LDL-P, when assessing CVD risk in patients for whom PCSK9 inhibitors are considered as a lipid-lowering therapy. Numerous methods for measuring LDL subfractions and LDL-P are available, but several factors limit their availability. A lack of standardization makes comparison between the different methods challenging. Longitudinal population-based studies have found an independent association between different LDL subfractions, LDL-P, and an increased risk of cardiovascular events, but definitive evidence that these measurements add predictive value to the standard risk markers is lacking. No studies have proven that these measurements improve clinical outcomes. PCSK9 inhibitors seem to be effective at lowering all LDL subfractions and LDL-P, but any evidence that measuring LDL subfractions and LDL-P yield clinically useful information is lacking. Such analyses are currently not recommended when considering whether to initiate PCKS9 inhibitors in patients at risk of CVD.
Collapse
Affiliation(s)
| | - Anders Hovland
- Division of Internal Medicine, Nordland Hospital, N-8092 Bodø, Norway.
- Department of Clinical Medicine, University of Tromsø, N-9037 Tromsø, Norway.
| | - Knut Tore Lappegård
- Division of Internal Medicine, Nordland Hospital, N-8092 Bodø, Norway.
- Department of Clinical Medicine, University of Tromsø, N-9037 Tromsø, Norway.
| |
Collapse
|
32
|
Ciccarelli G, D'Elia S, De Paulis M, Golino P, Cimmino G. Lipid Target in Very High-Risk Cardiovascular Patients: Lesson from PCSK9 Monoclonal Antibodies. Diseases 2018; 6:diseases6010022. [PMID: 29562587 PMCID: PMC5871968 DOI: 10.3390/diseases6010022] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2018] [Revised: 03/12/2018] [Accepted: 03/14/2018] [Indexed: 12/11/2022] Open
Abstract
The role of low-density lipoproteins (LDLs) as a major risk factor for cardiovascular disease has been demonstrated by several epidemiological studies. The molecular basis for LDLs in atherosclerotic plaque formation and progression is not completely unraveled yet. Pharmacological modulation of plasma LDL-C concentrations and randomized clinical trials addressing the impact of lipid-lowering interventions on cardiovascular outcome have clearly shown that reducing plasma LDL-C concentrations results in a significant decrease in major cardiovascular events. For many years, statins have represented the most powerful pharmacological agents available to lower plasma LDL-C concentrations. In clinical trials, it has been shown that the greater the reduction in plasma LDL-C concentrations, the lower the rate of major cardiovascular events, especially in high-risk patients, because of multiple risk factors and recurrent events. However, in a substantial number of patients, the recommended LDL target is difficult to achieve because of different factors: genetic background (familial hypercholesterolemia), side effects (statin intolerance), or high baseline plasma LDL-C concentrations. In the last decade, our understanding of the molecular mechanisms involved in LDL metabolism has progressed significantly and the key role of proprotein convertase subtilisin/kexin type 9 (PCSK9) has emerged. This protein is an enzyme able to bind the LDL receptors (LDL-R) on hepatocytes, favoring their degradation. Blocking PCSK9 represents an intriguing new therapeutic approach to decrease plasma LDL-C concentrations, which in recent studies has been demonstrated to also result in a significant reduction in major cardiovascular events.
Collapse
Affiliation(s)
- Giovanni Ciccarelli
- Department of Cardio-Thoracic and Respiratory Sciences, Section of Cardiology, University of Campania "Luigi Vanvitelli", 80131 Naples, Italy.
| | - Saverio D'Elia
- Department of Cardio-Thoracic and Respiratory Sciences, Section of Cardiology, University of Campania "Luigi Vanvitelli", 80131 Naples, Italy.
| | - Michele De Paulis
- Department of Cardio-Thoracic and Respiratory Sciences, Section of Cardiology, University of Campania "Luigi Vanvitelli", 80131 Naples, Italy.
| | - Paolo Golino
- Department of Cardio-Thoracic and Respiratory Sciences, Section of Cardiology, University of Campania "Luigi Vanvitelli", 80131 Naples, Italy.
| | - Giovanni Cimmino
- Department of Cardio-Thoracic and Respiratory Sciences, Section of Cardiology, University of Campania "Luigi Vanvitelli", 80131 Naples, Italy.
| |
Collapse
|
33
|
Abstract
INTRODUCTION Dyslipidemia is one of the most important risk factors for cardiovascular disease. Insufficient reduction in LDL-C from existing therapies in patients at high risk of atherogenic cardiovascular disease is an unmet clinical need. Circulating PCSK9 causes hypercholesterolemia by reducing LDL receptors in hepatocytes. Areas covered: PCSK9 inhibition has emerged as a promising new therapeutic strategy to reduce LDL-C. Inclisiran, a novel, synthetic, siRNA molecule, inhibits PCSK9 synthesis in hepatocytes. Inclisiran targets intracellular PCSK9 synthesis specifically, resulting in a dose-dependent, long-term, significant reduction in LDL-C. Inclisiran has been well tolerated and safe, without severe adverse events so far. This review discusses current PCSK9 inhibitors and the results of phase I and II clinical trials of inclisiran. Expert opinion: Plasma PCSK9 enhances the degradation of LDL receptor, resulting in accumulation of LDL-C in the circulation. Current approaches with monoclonal antibodies sequester circulating PCSK9 but require frequent injections. Inclisiran inhibits translation of PCSK9 mRNA and thus switches off PCSK9 production and provides advantages over monoclonal antibodies with an infrequent dosing interval of twice a year to reduce LDL-C by over 50%. Ongoing studies will establish the long-term safety of inclisiran in patients with high cardiovascular risk and an elevated LDL-C.
Collapse
Affiliation(s)
- Toshiyuki Nishikido
- a Imperial Centre for Cardiovascular Disease Prevention (ICCP), Department of Primary Care and Public Health , School of Public Health, Imperial College London , UK.,b Department of cardiovascular medicine , Saga University , Saga , Japan
| | - Kausik K Ray
- a Imperial Centre for Cardiovascular Disease Prevention (ICCP), Department of Primary Care and Public Health , School of Public Health, Imperial College London , UK
| |
Collapse
|
34
|
Baragetti A, Grejtakova D, Casula M, Olmastroni E, Jotti GS, Norata GD, Catapano AL, Bellosta S. Proprotein Convertase Subtilisin-Kexin type-9 (PCSK9) and triglyceride-rich lipoprotein metabolism: Facts and gaps. Pharmacol Res 2018; 130:1-11. [PMID: 29428206 DOI: 10.1016/j.phrs.2018.01.025] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2017] [Revised: 01/24/2018] [Accepted: 01/26/2018] [Indexed: 01/24/2023]
Abstract
After more than a decade of intense investigation, Pro-protein Convertase Subtilisin-Kexin type 9 (PCSK9) remains a hot topic of research both at experimental and clinical level. Interestingly PCSK9 is expressed in different tissues suggesting the existence of additional function(s) beyond the modulation of the Low-Density Lipoprotein (LDL) receptor in the liver. Emerging data suggest that PCSK9 might play a role in the modulation of triglyceride-rich lipoprotein (TGRL) metabolism, mainly Very Low-Density Lipoproteins (VLDL) and their remnants. In vitro, PCSK9 affects TGRLs production by intestinal cells as well as the catabolism of LDL receptor homologous and non-homologous targets such as VLDL receptor, CD36 and ApoE2R. However, the in vivo relevance of these findings is still debated. This review aims at critically discussing the role of PCSK9 on TGRLs metabolism with a major focus on the impact of its genetic and pharmacological modulation on circulating lipids and lipoproteins beyond LDL.
Collapse
Affiliation(s)
- Andrea Baragetti
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, via Balzaretti 9, 20133, Milan, Italy; S.I.S.A. Center for the Study of Atherosclerosis - Bassini Hospital, Cinisello Balsamo, Milan, Italy
| | | | - Manuela Casula
- Epidemiology and Preventive Pharmacology Centre (SEFAP), Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Via Balzaretti, 9, 20133 Milano, Italy
| | - Elena Olmastroni
- Epidemiology and Preventive Pharmacology Centre (SEFAP), Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Via Balzaretti, 9, 20133 Milano, Italy
| | - Gloria Saccani Jotti
- Department of Medicine & Surgery, Faculty of Medicine, University of Parma, Via Volturno 39, 43121 Parma, Italy
| | - Giuseppe Danilo Norata
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, via Balzaretti 9, 20133, Milan, Italy; School of Biomedical Sciences, Curtin Health Innovation Research Institute, Curtin University, Perth, Kent St., Bentley Western Australia 6102, Australia
| | - Alberico L Catapano
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, via Balzaretti 9, 20133, Milan, Italy; IRCCS MultiMedica, via Fantoli 16, 20138, Milan, Italy.
| | - Stefano Bellosta
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, via Balzaretti 9, 20133, Milan, Italy; IRCCS MultiMedica, via Fantoli 16, 20138, Milan, Italy
| |
Collapse
|
35
|
Peripheral vascular atherosclerosis in a novel PCSK9 gain-of-function mutant Ossabaw miniature pig model. Transl Res 2018; 192:30-45. [PMID: 29175268 PMCID: PMC5811343 DOI: 10.1016/j.trsl.2017.10.007] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/23/2016] [Revised: 10/18/2017] [Accepted: 10/24/2017] [Indexed: 10/24/2022]
Abstract
Hypercholesterolemia is a major risk factor for atherosclerosis. Remaining challenges in the management of atherosclerosis necessitate development of animal models that mimic human pathophysiology. We characterized a novel mutant pig model with DNA transposition of D374Y gain-of-function (GOF) cDNA of chimp proprotein convertase subtilisin/kexin type-9 (PCSK9), and tested the hypothesis that it would develop peripheral vascular remodeling and target organ injury in the kidney. Wild-type or PCSK9-GOF Ossabaw miniature pigs fed a standard or atherogenic diet (AD) (n = 7 each) were studied in vivo after 3 and 6 months of diet. Single-kidney hemodynamics and function were studied using multidetector computed tomography and kidney oxygenation by blood oxygen level-dependent magnetic resonance imaging. The renal artery was evaluated by intravascular ultrasound, aortic stiffness by multidetector computed tomography, and kidney stiffness by magnetic resonance elastography. Subsequent ex vivo studies included the renal artery endothelial function and morphology of abdominal aorta, renal, and femoral arteries by histology. Compared with wild type, PCSK9-GOF pigs had elevated cholesterol, triglyceride, and blood pressure levels at 3 and 6 months. Kidney stiffness increased in GOF groups, but aortic stiffness only in GOF-AD. Hypoxia, intrarenal fat deposition, oxidative stress, and fibrosis were observed in both GOF groups, whereas kidney function remained unchanged. Peripheral arteries in GOF groups showed medial thickening and development of atheromatous plaques. Renal endothelial function was impaired only in GOF-AD. Therefore, the PCSK9-GOF mutation induces rapid development of atherosclerosis in peripheral vessels of Ossabaw pigs, which is exacerbated by a high-cholesterol diet. This model may be useful for preclinical studies of atherosclerosis.
Collapse
|
36
|
Filippatos TD, Kei A, Rizos CV, Elisaf MS. Effects of PCSK9 Inhibitors on Other than Low-Density Lipoprotein Cholesterol Lipid Variables. J Cardiovasc Pharmacol Ther 2017; 23:3-12. [DOI: 10.1177/1074248417724868] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Low-density lipoprotein cholesterol (LDL-C) is a major cardiovascular risk factor, but other lipid variables such as triglycerides (TRGs), high-density lipoprotein cholesterol (HDL-C) and lipoprotein a [Lp(a)] also affect cardiovascular risk. Proprotein convertase subtilisin/kexin type 9 (PCSK9) inhibitors significantly lower LDL-C concentration but also modestly improve the concentrations of TRGs and HDL-C and more robustly decrease Lp(a) levels. The review presents the associated mechanisms of the beneficial effects of PCSK9 inhibitors on the other than LDL-C lipid variables, including the effects on lipid/apolipoprotein secretion and clearance and the heteroexchange between lipoproteins, as well as the possible effects on other variables involved in lipid metabolism such as sortilin. Proprotein convertase subtilisin/kexin type 9 inhibitors improve the overall lipid profile, and these beneficial effects may play a role in the reduction of cardiovascular risk.
Collapse
Affiliation(s)
| | - Anastazia Kei
- Department of Internal Medicine, School of Medicine, University of Ioannina, Ioannina, Greece
| | - Christos V. Rizos
- Department of Internal Medicine, School of Medicine, University of Ioannina, Ioannina, Greece
| | - Moses S. Elisaf
- Department of Internal Medicine, School of Medicine, University of Ioannina, Ioannina, Greece
| |
Collapse
|
37
|
Eisenga MF, Zelle DM, Sloan JH, Gaillard CAJM, Bakker SJL, Dullaart RPF. High Serum PCSK9 Is Associated With Increased Risk of New-Onset Diabetes After Transplantation in Renal Transplant Recipients. Diabetes Care 2017; 40:894-901. [PMID: 28461454 DOI: 10.2337/dc16-2258] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/21/2016] [Accepted: 04/06/2017] [Indexed: 02/03/2023]
Abstract
OBJECTIVE New-onset diabetes after transplantation (NODAT) is a major complication in renal transplant recipients (RTRs). Cholesterol metabolism has been linked to diabetes development. Proprotein convertase subtilisin/kexin type 9 (PCSK9) is crucial in LDL receptor regulation. Its association with NODAT is unknown. We prospectively determined the association between serum PCSK9 levels and NODAT development and then with all-cause mortality, cardiovascular mortality, and renal graft failure. RESEARCH DESIGN AND METHODS In a university setting, nondiabetic RTRs recruited between 2001 and 2003 with a functional graft for ≥1 year were eligible. Serum PCSK9 was measured by ELISA. Cox proportional hazards analysis was used to assess the association of PCSK9 with the development of NODAT, all-cause mortality, cardiovascular mortality, and graft failure. RESULTS In 453 RTRs (age 51 ± 12 years, 56% male; 6.1 [2.7-11.7] years after transplantation), serum PCSK9 was 107.1 ± 43.4 μg/L. During a median follow-up of 10 years, 70 RTRs developed NODAT, 123 died, and 59 developed graft failure. NODAT occurred more frequently in the upper PCSK9 tertile (23%) versus the lowest two PCSK9 tertiles (12%; P < 0.001). In crude Cox regression analyses, PCSK9 was significantly associated with development of NODAT (hazard ratio 1.34 [95% CI 1.10-1.63]) per SD change (P = 0.004). This association remained independent of adjustment for potential confounders, including statin use. PCSK9 was not associated with all-cause mortality, cardiovascular mortality, or graft failure. CONCLUSIONS Circulating PCSK9 is associated with NODAT in RTRs. The PCSK9 pathway may contribute to the pathogenesis of NODAT.
Collapse
Affiliation(s)
- Michele F Eisenga
- Department of Internal Medicine, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Dorien M Zelle
- Department of Internal Medicine, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - John H Sloan
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN
| | - Carlo A J M Gaillard
- Department of Internal Medicine, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Stephan J L Bakker
- Department of Internal Medicine, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Robin P F Dullaart
- Department of Internal Medicine, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| |
Collapse
|
38
|
Murphy BA, Tadin-Strapps M, Jensen K, Mogg R, Liaw A, Herath K, Bhat G, McLaren DG, Previs SF, Pinto S. siRNA-mediated inhibition of SREBP cleavage-activating protein reduces dyslipidemia in spontaneously dysmetabolic rhesus monkeys. Metabolism 2017; 71:202-212. [PMID: 28521874 DOI: 10.1016/j.metabol.2017.02.015] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/13/2016] [Revised: 02/26/2017] [Accepted: 02/27/2017] [Indexed: 01/07/2023]
Abstract
BACKGROUND SREBP cleavage-activating protein (SCAP) is a cholesterol binding endoplasmic reticulum (ER) membrane protein that is required to activate SREBP transcription factors. SREBPs regulate genes involved in lipid biosynthesis. They also influence lipid clearance by modulating the expression of LDL receptor (LDLR) and proprotein convertase subtilisin/kexin type 9 (PCSK9) genes. Inhibiting SCAP decreases circulating PCSK9, triglycerides (TG), and LDL-cholesterol (LDL-C), both in vitro and in vivo. Type 2 diabetics with dyslipidemia are at high risk for cardiovascular diseases. These patients present a unique pathophysiological lipid profile characterized by moderately elevated LDL-C, elevated TG and reduced HDL-cholesterol (HDL-C). The spontaneous dysmetabolic rhesus monkey model (DysMet RhM) recapitulates this human dyslipidemia and therefore is an attractive preclinical model to evaluate SCAP inhibition as a therapy for this disease population. The objective to of this study was to assess the effect of SCAP inhibition on the lipid profile of DysMet RhM. METHOD We assessed the effect of inhibiting hepatic SCAP on the lipid profile of DysMet RhM using an siRNA encapsulated lipid nanoparticle (siRNA-LNP). RESULTS The SCAP siRNA-LNP significantly reduced LDL-C, PCSK9 and TG in DysMet RhM; LDL-C was reduced by ≥20%, circulating PCSK9 by 30-40% and TG by >25%. These changes by the SCAP siRNA-LNP agree with the predicted effect of SCAP inhibition and reduced SREBP tone on these endpoints. CONCLUSION These data demonstrate that a SCAP siRNA-LNP improved the lipid profile in a clinically relevant preclinical disease model and provide evidence for SCAP inhibition as a therapy for diabetic dyslipidemic patients.
Collapse
Affiliation(s)
- Beth Ann Murphy
- Pharmacology, Merck &Co. Inc., 2000 Galloping Hill Rd., Kenilworth, NJ 07033, USA.
| | - Marija Tadin-Strapps
- Genetics and Pharmacogenomics, Merck & Co. Inc., 33 Avenue Louis Pasteur, Boston, MA 02115, USA
| | - Kristian Jensen
- Cardiometabolic Disease, Merck & Co. Inc., 2000 Galloping Hill Rd., Kenilworth, NJ 07033, USA
| | - Robin Mogg
- Biostatistics, Merck & Co. Inc., 351 North Sumneytown Pike, North Wales, PA 19454, USA
| | - Andy Liaw
- Biostatistics, Merck & Co. Inc., 126 E. Lincoln Avenue, PO Box 2000, Rahway, NJ 07065, USA
| | - Kithsiri Herath
- Cardiometabolic Disease, Merck & Co. Inc., 2000 Galloping Hill Rd., Kenilworth, NJ 07033, USA
| | - Gowri Bhat
- Cardiometabolic Disease, Merck & Co. Inc., 2000 Galloping Hill Rd., Kenilworth, NJ 07033, USA
| | - David G McLaren
- Pharmacology, Merck &Co. Inc., 2000 Galloping Hill Rd., Kenilworth, NJ 07033, USA
| | - Stephen F Previs
- Cardiometabolic Disease, Merck & Co. Inc., 2000 Galloping Hill Rd., Kenilworth, NJ 07033, USA
| | - Shirly Pinto
- Cardiometabolic Disease, Merck & Co. Inc., 2000 Galloping Hill Rd., Kenilworth, NJ 07033, USA
| |
Collapse
|
39
|
Affiliation(s)
- Robin P F Dullaart
- From the Department of Endocrinology, University of Groningen, University Medical Center, Groningen, the Netherlands
| |
Collapse
|
40
|
Abstract
Even though it is only a little over a decade from the discovery of proprotein convertase subtilisin/kexin type 9 (PCSK9) as a plasma protein that associates with both high and low cholesterol syndromes, a rich body of knowledge has developed, and drugs inhibiting this target have been approved in many markets. While the majority of research in recent years has focused on the impact of therapeutic antagonism of this molecule, important lines of investigation have emerged characterizing its unique physiology as it relates to cholesterol metabolism and atherosclerosis. The PCSK9 story is unfolding rapidly but is far from complete. One chapter that is of particular interest is the possible direct link between PCSK9 and atherosclerosis. This review specifically examines this relationship drawing from data produced from experimental models of plaque biology and inflammation, atherosclerosis imaging studies, and observational epidemiology.
Collapse
Affiliation(s)
- Michael D Shapiro
- Oregon Health & Science University, Knight Cardiovascular Institute, Center for Preventive Cardiology
| | - Sergio Fazio
- Oregon Health & Science University, Knight Cardiovascular Institute, Center for Preventive Cardiology
| |
Collapse
|
41
|
Viecili PRN, da Silva B, Hirsch GE, Porto FG, Parisi MM, Castanho AR, Wender M, Klafke JZ. Triglycerides Revisited to the Serial. Adv Clin Chem 2017; 80:1-44. [PMID: 28431638 DOI: 10.1016/bs.acc.2016.11.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
This review discusses the role of triglycerides (TGs) in the normal cardiovascular system as well as in the development and clinical manifestation of cardiovascular diseases. Regulation of TGs at the enzymatic and genetic level, in addition to their possible relevance as preclinical and clinical biomarkers, is discussed, culminating with a description of available and emerging treatments. Due to the high complexity of the subject and the vast amount of material in the literature, the objective of this review was not to exhaust the subject, but rather to compile the information to facilitate and improve the understanding of those interested in this topic. The main publications on the topic were sought out, especially those from the last 5 years. The data in the literature still give reason to believe that there is room for doubt regarding the use of TG as disease biomarkers; however, there is increasing evidence for the role of hypertriglyceridemia on the atherosclerotic inflammatory process, cardiovascular outcomes, and mortality.
Collapse
|
42
|
Ferri N, Ruscica M. Proprotein convertase subtilisin/kexin type 9 (PCSK9) and metabolic syndrome: insights on insulin resistance, inflammation, and atherogenic dyslipidemia. Endocrine 2016; 54:588-601. [PMID: 27038318 DOI: 10.1007/s12020-016-0939-0] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/21/2016] [Accepted: 03/22/2016] [Indexed: 02/07/2023]
Abstract
Low-density lipoprotein (LDL) cholesterol plays a pivotal role in the pathogenesis of atherosclerotic cardiovascular disease (CVD). The discovery that proprotein convertase subtilisin/kexin type 9 (PCSK9) represents a key regulator pathway for hepatic LDL receptor (LDLR) degradation sheds light on new uncovered issues regarding LDL-C homeostasis. Indeed, as confirmed by phase II and III clinical trials with monoclonal antibodies, targeting PCSK9 represents the newest and most promising pharmacological tool for the treatment of hypercholesterolemia and related CVD. However, clinical, genetic, and experimental evidence indicates that PCSK9 may be either a cause or an effect in the context of metabolic syndrome (MetS), a condition comprising a cluster of risk factors including insulin resistance, obesity, hypertension, and atherogenic dyslipidemia. The latter is characterized by a triad of hypertriglyceridemia, low plasma concentrations of high-density lipoproteins, and qualitative changes in LDLs. PCSK9 levels seem to correlate with many of these lipid parameters as well as with the insulin sensitivity indices, although the molecular mechanisms behind this association are still unknown or not completely elucidated. Nevertheless, this area of research represents an important starting point for a better understanding of the physiological role of PCSK9, also considering the recent approval of new therapies involving anti-PCSK9. Thus, in the present review, we will discuss the current knowledge on the role of PCSK9 in the context of MetS, alteration of lipids, glucose homeostasis, and inflammation.
Collapse
Affiliation(s)
- Nicola Ferri
- Dipartimento di Scienze del Farmaco, Università degli Studi di Padova, Largo Meneghetti 2, 35131, Padua, Italy
| | - Massimiliano Ruscica
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Via Balzaretti 9, 20133, Milan, Italy.
| |
Collapse
|
43
|
Verbeek R, Boyer M, Boekholdt SM, Hovingh GK, Kastelein JJP, Wareham N, Khaw KT, Arsenault BJ. Carriers of the PCSK9 R46L Variant Are Characterized by an Antiatherogenic Lipoprotein Profile Assessed by Nuclear Magnetic Resonance Spectroscopy-Brief Report. Arterioscler Thromb Vasc Biol 2016; 37:43-48. [PMID: 27856457 DOI: 10.1161/atvbaha.116.307995] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2016] [Accepted: 10/17/2016] [Indexed: 11/16/2022]
Abstract
OBJECTIVE Carriers of the PCSK9 (proprotein convertase subtilisin/kexin 9) R46L genetic variant (rs11591147) are characterized by low levels of low-density lipoprotein cholesterol and a decreased risk of cardiovascular disease. We studied the impact of the R46L variant on lipoprotein size and composition. APPROACH AND RESULTS Lipoprotein size and composition were measured by nuclear magnetic resonance spectroscopy in 2373 participants of the EPIC (European Prospective Investigation into Cancer and Nutrition)-Norfolk study. After adjusting for age, sex, and cardiovascular disease status, carriers of the R46L variant (n=77) were characterized by lower concentrations of very low-density lipoprotein particles (85.8±26.2 versus 99.0±33.3 nmol/L; P<0.001), low-density lipoprotein particles (1479.7±396.8 versus 1662.9±458.3 nmol/L; P<0.001), and lipoprotein(a) (11.1 [7.2-28.6] versus 12.4 [6.7-29.1] mg/dL; P<0.001) compared with noncarriers. Total high-density lipoprotein particle and very low-density lipoprotein, low-density lipoprotein, and high-density lipoprotein particle sizes were comparable in carriers and noncarriers. Carriers were characterized by lower secretory phospholipase A2 (4.2±0.9 versus 4.6±1.3 nmol/mL/min; P=0.004) and lipoprotein-associated phospholipase A2 activity (47.5±14.1 versus 52.4±16.2 nmol/mL/min; P=0.02) compared with noncarriers. CONCLUSIONS Results of this study suggest that carriers of the PCSK9 R46L genetic variant have lower very low-density lipoprotein and low-density lipoprotein particle concentrations, lower lipoprotein(a) levels, and lower secretory phospholipase A2 and lipoprotein-associated phospholipase A2 activity compared with noncarriers.
Collapse
Affiliation(s)
- Rutger Verbeek
- From the Department of Vascular Medicine (R.V., G.K.H., J.J.P.K.) and Department of Cardiology (S.M.B.), Academic Medical Centre, Amsterdam, The Netherlands; Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec, Canada (M.B., B.J.A.); Department of Medicine, Faculty of Medicine, Université Laval, Québec, Canada (M.B., B.J.A.); Medical Research Council Epidemiology Unit, Cambridge, United Kingdom (N.W.); and Department of Public Health and Primary Care, Institute of Public Health, University of Cambridge, United Kingdom (K.-T.K.)
| | - Marjorie Boyer
- From the Department of Vascular Medicine (R.V., G.K.H., J.J.P.K.) and Department of Cardiology (S.M.B.), Academic Medical Centre, Amsterdam, The Netherlands; Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec, Canada (M.B., B.J.A.); Department of Medicine, Faculty of Medicine, Université Laval, Québec, Canada (M.B., B.J.A.); Medical Research Council Epidemiology Unit, Cambridge, United Kingdom (N.W.); and Department of Public Health and Primary Care, Institute of Public Health, University of Cambridge, United Kingdom (K.-T.K.)
| | - S Matthijs Boekholdt
- From the Department of Vascular Medicine (R.V., G.K.H., J.J.P.K.) and Department of Cardiology (S.M.B.), Academic Medical Centre, Amsterdam, The Netherlands; Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec, Canada (M.B., B.J.A.); Department of Medicine, Faculty of Medicine, Université Laval, Québec, Canada (M.B., B.J.A.); Medical Research Council Epidemiology Unit, Cambridge, United Kingdom (N.W.); and Department of Public Health and Primary Care, Institute of Public Health, University of Cambridge, United Kingdom (K.-T.K.)
| | - G Kees Hovingh
- From the Department of Vascular Medicine (R.V., G.K.H., J.J.P.K.) and Department of Cardiology (S.M.B.), Academic Medical Centre, Amsterdam, The Netherlands; Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec, Canada (M.B., B.J.A.); Department of Medicine, Faculty of Medicine, Université Laval, Québec, Canada (M.B., B.J.A.); Medical Research Council Epidemiology Unit, Cambridge, United Kingdom (N.W.); and Department of Public Health and Primary Care, Institute of Public Health, University of Cambridge, United Kingdom (K.-T.K.)
| | - John J P Kastelein
- From the Department of Vascular Medicine (R.V., G.K.H., J.J.P.K.) and Department of Cardiology (S.M.B.), Academic Medical Centre, Amsterdam, The Netherlands; Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec, Canada (M.B., B.J.A.); Department of Medicine, Faculty of Medicine, Université Laval, Québec, Canada (M.B., B.J.A.); Medical Research Council Epidemiology Unit, Cambridge, United Kingdom (N.W.); and Department of Public Health and Primary Care, Institute of Public Health, University of Cambridge, United Kingdom (K.-T.K.)
| | - Nicholas Wareham
- From the Department of Vascular Medicine (R.V., G.K.H., J.J.P.K.) and Department of Cardiology (S.M.B.), Academic Medical Centre, Amsterdam, The Netherlands; Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec, Canada (M.B., B.J.A.); Department of Medicine, Faculty of Medicine, Université Laval, Québec, Canada (M.B., B.J.A.); Medical Research Council Epidemiology Unit, Cambridge, United Kingdom (N.W.); and Department of Public Health and Primary Care, Institute of Public Health, University of Cambridge, United Kingdom (K.-T.K.)
| | - Kay-Tee Khaw
- From the Department of Vascular Medicine (R.V., G.K.H., J.J.P.K.) and Department of Cardiology (S.M.B.), Academic Medical Centre, Amsterdam, The Netherlands; Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec, Canada (M.B., B.J.A.); Department of Medicine, Faculty of Medicine, Université Laval, Québec, Canada (M.B., B.J.A.); Medical Research Council Epidemiology Unit, Cambridge, United Kingdom (N.W.); and Department of Public Health and Primary Care, Institute of Public Health, University of Cambridge, United Kingdom (K.-T.K.)
| | - Benoit J Arsenault
- From the Department of Vascular Medicine (R.V., G.K.H., J.J.P.K.) and Department of Cardiology (S.M.B.), Academic Medical Centre, Amsterdam, The Netherlands; Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec, Canada (M.B., B.J.A.); Department of Medicine, Faculty of Medicine, Université Laval, Québec, Canada (M.B., B.J.A.); Medical Research Council Epidemiology Unit, Cambridge, United Kingdom (N.W.); and Department of Public Health and Primary Care, Institute of Public Health, University of Cambridge, United Kingdom (K.-T.K.).
| |
Collapse
|
44
|
Nozue T, Hattori H, Ogawa K, Kujiraoka T, Iwasaki T, Hirano T, Michishita I. Correlation between serum levels of proprotein convertase subtilisin/kexin type 9 (PCSK9) and atherogenic lipoproteins in patients with coronary artery disease. Lipids Health Dis 2016; 15:165. [PMID: 27658826 PMCID: PMC5034502 DOI: 10.1186/s12944-016-0339-8] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2016] [Accepted: 09/17/2016] [Indexed: 02/06/2023] Open
Abstract
Background Proprotein convertase subtilisin/kexin type 9 (PCSK9) is a key regulator of serum low-density lipoprotein (LDL) cholesterol levels. Recently, PCSK9 has additionally been related to metabolic risk factors such as the levels of triglycerides, apolipoprotein B (apoB), insulin, and glucose, as well as body mass index. The purpose of this study was to investigate correlations between serum levels of PCSK9 and apoB-containing atherogenic lipoproteins in patients with coronary artery disease (CAD). Methods Serum levels of PCSK9 and lipoprotein(a) [Lp(a)]; small, dense LDL; and oxidized LDL were measured in 101 patients with CAD who were not receiving lipid-lowering therapy. Results Serum hetero-dimer PCSK9 levels were positively correlated with serum levels of Lp(a) (r = 0.195, p = 0.05); small, dense LDL (r = 0.336, p = 0.0006); and oxidized LDL (r = 0.268, p = 0.008). Multivariate regression analyses showed that serum hetero-dimer PCSK9 was a significant predictor of serum levels of Lp(a) (β = 0.235, p = 0.01); small, dense LDL (β = 0.143, p = 0.03); and oxidized LDL (β = 0.268, p = 0.008). Conclusions Serum PCSK9 levels were positively correlated with serum levels of Lp(a); small, dense LDL; and oxidized LDL in patients with CAD. This suggests that the interaction between serum PCSK9 and apoB-containing lipoproteins plays a role in establishing the atherosclerotic status of patients. Trial registration UMIN Clinical Trials Registry, UMIN ID: C000000311.
Collapse
Affiliation(s)
- Tsuyoshi Nozue
- Division of Cardiology, Department of Internal Medicine, Yokohama Sakae Kyosai Hospital, Federation of National Public Service Personnel Mutual Associations, 132 Katsura-cho, Sakae-ku, Yokohama, 247-8581, Japan.
| | - Hiroaki Hattori
- Advanced Medical Technology and Development Division, BML Inc., Kawagoe, Japan
| | - Kazuyuki Ogawa
- Advanced Medical Technology and Development Division, BML Inc., Kawagoe, Japan
| | - Takeshi Kujiraoka
- Advanced Medical Technology and Development Division, BML Inc., Kawagoe, Japan
| | - Tadao Iwasaki
- Advanced Medical Technology and Development Division, BML Inc., Kawagoe, Japan
| | - Tsutomu Hirano
- Department of Medicine, Division of Diabetes, Metabolism, and Endocrinology, Showa University School of Medicine, Tokyo, Japan
| | - Ichiro Michishita
- Division of Cardiology, Department of Internal Medicine, Yokohama Sakae Kyosai Hospital, Federation of National Public Service Personnel Mutual Associations, 132 Katsura-cho, Sakae-ku, Yokohama, 247-8581, Japan
| |
Collapse
|
45
|
Norata GD, Tavori H, Pirillo A, Fazio S, Catapano AL. Biology of proprotein convertase subtilisin kexin 9: beyond low-density lipoprotein cholesterol lowering. Cardiovasc Res 2016; 112:429-42. [PMID: 27496869 DOI: 10.1093/cvr/cvw194] [Citation(s) in RCA: 104] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/05/2016] [Accepted: 07/06/2016] [Indexed: 12/17/2022] Open
Abstract
Proprotein convertase subtilisin kexin 9 (PCSK9) is a key regulator of low-density lipoprotein receptor levels and LDL-cholesterol levels. Loss-of-function mutations in PCSK9 gene are associated with hypocholesterolaemia and protection against cardiovascular disease, identifying PCSK9 inhibition as a valid therapeutic approach to manage hypercholesterolaemia and related diseases. Although PCSK9 is expressed mainly in the liver, it is present also in other tissues and organs with specific functions, raising the question of whether a pharmacological inhibition of PCSK9 to treat hypercholesterolaemia and associated cardiovascular diseases might be helpful or deleterious in non-hepatic tissues. For example, PCSK9 is expressed in the vascular wall, in the kidneys, and in the brain, where it was proposed to play a role in development, neurocognitive process, and neuronal apoptosis. A link between PCSK9 and immunity was also proposed as both sepsis and viral infections are differentially affected in the presence or absence of PCSK9. Despite the increasing number of observations, the debate on the exact roles of PCSK9 in extrahepatic tissues is still ongoing, and as very effective drugs that inhibit PCSK9 have become available to the clinician, a better understanding of the biological roles of PCSK9 is warranted.
Collapse
Affiliation(s)
- Giuseppe Danilo Norata
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milan, Italy Center for the Study of Atherosclerosis, Ospedale Bassini, Cinisello Balsamo, Italy
| | - Hagai Tavori
- Center for Preventive Cardiology, Knight Cardiovascular Institute, Oregon Health & Science University, Portland, OR, USA
| | - Angela Pirillo
- Center for the Study of Atherosclerosis, Ospedale Bassini, Cinisello Balsamo, Italy IRCCS Multimedica, Milan, Italy
| | - Sergio Fazio
- Center for Preventive Cardiology, Knight Cardiovascular Institute, Oregon Health & Science University, Portland, OR, USA
| | - Alberico L Catapano
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milan, Italy IRCCS Multimedica, Milan, Italy
| |
Collapse
|
46
|
Abstract
PURPOSE OF REVIEW High levels of LDL-cholesterol (LDL-C) are directly associated with devastating cardiovascular complications. Statins downregulate cholesterol synthesis and upregulate hepatic mRNA levels of LDL receptor (LDLR) and proprotein convertase subtilisin-kexin 9 (PCSK9), a validated enhancer of LDLR protein degradation. Herein, we summarize recent discoveries of the biological properties of PCSK9 in both health and disease states. RECENT FINDINGS PCSK9 downregulation of the LDLR protein likely explains the observed protective effect of the loss of PCSK9 in reducing lipoprotein(a) and incidence of septic shock. Injectable inhibitory PCSK9 monoclonal antibodies are now prescribed to hypercholesterolemic patients that do not reach target levels of LDL-C with available drugs. PCSK9 also reduces the levels of other receptors, for example, VLDL receptor (VLDLR), ApoER2, CD36, and CD81. The efficacy of the upregulation of LDLR and VLDLR cell surface levels in the absence of PCSK9 is both tissue and sex dependent. As LDLR, CD81, and VLDLR are hepatitis C receptors, PCSK9 may protect against certain viral infections. SUMMARY New functions of PCSK9 and other receptor targets are beginning to emerge to explain the observed changes in LDL-C and triglycerides. The effect of PCSK9 loss-of-function on glucose metabolism, factors that regulate the expression of PCSK9, and the roles of PCSK9 in other tissues, for example, intestine, kidney, and brain require further investigations.
Collapse
Affiliation(s)
- Nabil G Seidah
- Institut de Recherches Cliniques de Montréal (IRCM), Affiliated with the Université de Montréal, Montreal, Québec, Canada
| |
Collapse
|
47
|
Walley KR, Francis GA, Opal SM, Stein EA, Russell JA, Boyd JH. The Central Role of Proprotein Convertase Subtilisin/Kexin Type 9 in Septic Pathogen Lipid Transport and Clearance. Am J Respir Crit Care Med 2016; 192:1275-86. [PMID: 26252194 DOI: 10.1164/rccm.201505-0876ci] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Microbial cell walls contain pathogenic lipids, including LPS in gram-negative bacteria, lipoteichoic acid in gram-positive bacteria, and phospholipomannan in fungi. These pathogen lipids are major ligands for innate immune receptors and figure prominently in triggering the septic inflammatory response. Alternatively, pathogen lipids can be cleared and inactivated, thus limiting the inflammatory response. Accordingly, biological mechanisms for sequestering and clearing pathogen lipids from the circulation have evolved. Pathogen lipids released into the circulation are initially bound by transfer proteins, notably LPS binding protein and phospholipid transfer protein, and incorporated into high-density lipoprotein particles. Next, LPS binding protein, phospholipid transfer protein, and other transfer proteins transfer these lipids to ApoB-containing lipoproteins, including low-density (LDL) and very-low-density lipoproteins and chylomicrons. Pathogen lipids within these lipoproteins and their remnants are then cleared from the circulation by the liver. Hepatic clearance involves the LDL receptor (LDLR) and possibly other receptors. Once absorbed by the liver, these lipids are then excreted in the bile. Recent evidence suggests pathogen lipid clearance can be modulated. Importantly, reduced proprotein convertase subtilisin/kexin type 9 activity increases recycling of the LDLR and thereby increases LDLR on the surface of hepatocytes, which increases clearance by the liver of pathogen lipids transported in LDL. Increased pathogen lipid clearance, which can be achieved by inhibiting proprotein convertase subtilisin/kexin type 9, may decrease the systemic inflammatory response to sepsis and improve clinical outcomes.
Collapse
Affiliation(s)
- Keith R Walley
- 1 Centre for Heart Lung Innovation, University of British Columbia, Vancouver, British Columbia, Canada
| | - Gordon A Francis
- 1 Centre for Heart Lung Innovation, University of British Columbia, Vancouver, British Columbia, Canada
| | - Steven M Opal
- 2 Infectious Disease Division, Memorial Hospital of Rhode Island and Alpert Medical School of Brown University, Providence, Rhode Island; and
| | - Evan A Stein
- 3 Metabolic and Atherosclerosis Research Center, Cincinnati, Ohio
| | - James A Russell
- 1 Centre for Heart Lung Innovation, University of British Columbia, Vancouver, British Columbia, Canada
| | - John H Boyd
- 1 Centre for Heart Lung Innovation, University of British Columbia, Vancouver, British Columbia, Canada
| |
Collapse
|
48
|
Xia CG, Zhang D, Ma C, Zhou J, He S, Su XR. Characterization and comparison of proteomes of albino sea cucumber Apostichopus japonicus (Selenka) by iTRAQ analysis. FISH & SHELLFISH IMMUNOLOGY 2016; 51:229-239. [PMID: 26707782 DOI: 10.1016/j.fsi.2015.12.027] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/03/2015] [Revised: 12/12/2015] [Accepted: 12/14/2015] [Indexed: 06/05/2023]
Abstract
Sea cucumber is a commercially important marine organism in China. Of the different colored varieties sold in China, albino sea cucumber has the greatest appeal among consumers. Identification of factors contributing to albinism in sea cucumber is therefore likely to provide a scientific basis for improving the cultivability of these strains. In this study, two-dimensional liquid chromatography-tandem mass spectrometry coupled with isobaric tags for relative and absolute quantification labeling was used for the first time to quantitatively define the proteome of sea cucumbers and reveal proteomic characteristics unique to albino sea cucumbers. A total of 549 proteins were identified and quantified in albino sea cucumber and the functional annotations of 485 proteins have been exhibited based on COG database. Compared with green sea cucumber, 12 proteins were identified as differentially expressed in the intestine and 16 proteins in the body wall of albino sea cucumber. Among them, 5 proteins were up-regulated in the intestine and 8 proteins were down-regulated in body wall. Gene ontology annotations of these differentially expressed proteins consisted mostly of 'biological process'. The large number of differentially expressed proteins identified here should be highly useful in further elucidating the mechanisms underlying albinism in sea cucumber.
Collapse
Affiliation(s)
- Chang-Ge Xia
- School of Marine Sciences, Ningbo University, Zhejiang Province 315211, PR China; Xinlicheng Reservoir Management Bureau in Changchun, Jilin Province 130119, PR China
| | - Dijun Zhang
- School of Marine Sciences, Ningbo University, Zhejiang Province 315211, PR China
| | - Chengnv Ma
- School of Marine Sciences, Ningbo University, Zhejiang Province 315211, PR China
| | - Jun Zhou
- School of Marine Sciences, Ningbo University, Zhejiang Province 315211, PR China
| | - Shan He
- School of Marine Sciences, Ningbo University, Zhejiang Province 315211, PR China
| | - Xiu-Rong Su
- School of Marine Sciences, Ningbo University, Zhejiang Province 315211, PR China.
| |
Collapse
|
49
|
Tavori H, Giunzioni I, Predazzi IM, Plubell D, Shivinsky A, Miles J, Devay RM, Liang H, Rashid S, Linton MF, Fazio S. Human PCSK9 promotes hepatic lipogenesis and atherosclerosis development via apoE- and LDLR-mediated mechanisms. Cardiovasc Res 2016; 110:268-78. [PMID: 26980204 DOI: 10.1093/cvr/cvw053] [Citation(s) in RCA: 76] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/21/2015] [Accepted: 03/08/2016] [Indexed: 01/07/2023] Open
Abstract
AIMS Proprotein convertase subtilisin/kexin type 9 (PCSK9) promotes the degradation of hepatic low-density lipoprotein (LDL) receptors (LDLR), thereby, decreasing hepatocyte LDL-cholesterol (LDL-C) uptake. However, it is unknown whether PCSK9 has effects on atherogenesis that are independent of lipid changes. The present study investigated the effect of human (h) PCSK9 on plasma lipids, hepatic lipogenesis, and atherosclerotic lesion size and composition in transgenic mice expressing hPCSK9 (hPCSK9tg) on wild-type (WT), LDLR⁻/⁻, or apoE⁻/⁻ background. METHODS AND RESULTS hPCSK9 expression significantly increased plasma cholesterol (+91%), triglycerides (+18%), and apoB (+57%) levels only in WT mice. The increase in plasma lipids was a consequence of both decreased hepatic LDLR and increased hepatic lipid production, mediated transcriptionally and post-transcriptionally by PCSK9 and dependent on both LDLR and apoE. Despite the lack of changes in plasma lipids in mice expressing hPCSK9 and lacking LDLR (the main target for PCSK9) or apoE (a canonical ligand for the LDLR), hPCSK9 expression increased aortic lesion size in the absence of apoE (268 655 ± 97 972 µm² in hPCSK9tg/apoE⁻/⁻ vs. 189 423 ± 65 700 µm(2) in apoE⁻/⁻) but not in the absence of LDLR. Additionally, hPCSK9 accumulated in the atheroma and increased lesion Ly6C(hi) monocytes (by 21%) in apoE⁻/⁻ mice, but not in LDLR⁻/⁻ mice. CONCLUSIONS PCSK9 increases hepatic lipid and lipoprotein production via apoE- and LDLR-dependent mechanisms. However, hPCSK9 also accumulate in the artery wall and directly affects atherosclerosis lesion size and composition independently of such plasma lipid and lipoprotein changes. These effects of hPCSK9 are dependent on LDLR but are independent of apoE.
Collapse
Affiliation(s)
- Hagai Tavori
- Knight Cardiovascular Institute, Center for Preventive Cardiology, Oregon Health and Science University, Portland, OR, USA
| | - Ilaria Giunzioni
- Knight Cardiovascular Institute, Center for Preventive Cardiology, Oregon Health and Science University, Portland, OR, USA
| | - Irene M Predazzi
- Knight Cardiovascular Institute, Center for Preventive Cardiology, Oregon Health and Science University, Portland, OR, USA
| | - Deanna Plubell
- Knight Cardiovascular Institute, Center for Preventive Cardiology, Oregon Health and Science University, Portland, OR, USA
| | - Anna Shivinsky
- Knight Cardiovascular Institute, Center for Preventive Cardiology, Oregon Health and Science University, Portland, OR, USA
| | - Joshua Miles
- Knight Cardiovascular Institute, Center for Preventive Cardiology, Oregon Health and Science University, Portland, OR, USA
| | - Rachel M Devay
- Rinat Laboratory, Pfizer Inc., South San Francisco, CA, USA
| | - Hong Liang
- Rinat Laboratory, Pfizer Inc., South San Francisco, CA, USA
| | - Shirya Rashid
- Department of Pharmacology, Dalhousie University, Halifax, Nova Scotia, Canada Dalhousie Medicine New Brunswick University, Saint John, New Brunswick, Canada
| | - MacRae F Linton
- Division of Cardiovascular Medicine, Atherosclerosis Research Unit, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Sergio Fazio
- Knight Cardiovascular Institute, Center for Preventive Cardiology, Oregon Health and Science University, Portland, OR, USA
| |
Collapse
|
50
|
Koren MJ, Kereiakes D, Pourfarzib R, Winegar D, Banerjee P, Hamon S, Hanotin C, McKenney JM. Effect of PCSK9 Inhibition by Alirocumab on Lipoprotein Particle Concentrations Determined by Nuclear Magnetic Resonance Spectroscopy. J Am Heart Assoc 2015; 4:e002224. [PMID: 26586732 PMCID: PMC4845239 DOI: 10.1161/jaha.115.002224] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/25/2015] [Accepted: 09/28/2015] [Indexed: 01/14/2023]
Abstract
BACKGROUND In patients with discordance between low-density lipoprotein (LDL) cholesterol and LDL particle (LDL-P) concentrations, cardiovascular risk more closely correlates with LDL-P. METHODS AND RESULTS We investigated the effect of alirocumab, a fully human monoclonal antibody to proprotein convertase subtilisin/kexin type 9, on lipoprotein particle concentration and size in hypercholesterolemic patients, using nuclear magnetic resonance spectroscopy. Plasma samples were collected from patients receiving alirocumab 150 mg every 2 weeks (n=26) or placebo (n=31) during a phase II, double-blind, placebo-controlled trial in patients (LDL cholesterol ≥100 mg/dL) on a stable atorvastatin dose. In this post hoc analysis, percentage change in concentrations of LDL-P, very-low-density lipoprotein particles, and high-density lipoprotein particles from baseline to week 12 was determined by nuclear magnetic resonance. Alirocumab significantly reduced mean concentrations of total LDL-P (-63.3% versus -1.0% with placebo) and large (-71.3% versus -21.8%) and small (-54.0% versus +17.8%) LDL-P subfractions and total very-low-density lipoprotein particle concentrations (-36.4% versus +33.4%; all P<0.01). Total high-density lipoprotein particles increased with alirocumab (+11.2% versus +1.4% with placebo; P<0.01). There were greater increases in large (44.6%) versus medium (17.7%) or small high-density lipoprotein particles (2.8%) with alirocumab. LDL-P size remained relatively unchanged in both groups; however, very-low-density and high-density lipoprotein particle sizes increased to a significantly greater extent with alirocumab. CONCLUSIONS Alirocumab significantly reduced LDL-C and LDL-P concentrations in hypercholesterolemic patients receiving stable atorvastatin therapy. These findings may be of particular relevance to patients with discordant LDL-C and LDL-P concentrations. CLINICAL TRIAL REGISTRATION URL: https://clinicaltrials.gov. Unique identifier: NCT01288443.
Collapse
Affiliation(s)
| | - Dean Kereiakes
- The Christ Hospital Heart and Vascular Center/The Lindner Research CenterCincinnatiOH
| | | | | | | | - Sara Hamon
- Regeneron Pharmaceuticals, IncTarrytownNY
| | | | - James M. McKenney
- Virginia Commonwealth University and National Clinical ResearchRichmondVA
| |
Collapse
|