1
|
Krishnamurthy G, Nguyen PT, Tran BN, Phan HT, Brennecke SP, Moses EK, Melton PE. Genomic variation associated with cardiovascular disease progression following preeclampsia: a systematic review. FRONTIERS IN EPIDEMIOLOGY 2023; 3:1221222. [PMID: 38455895 PMCID: PMC10911037 DOI: 10.3389/fepid.2023.1221222] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Accepted: 09/14/2023] [Indexed: 03/09/2024]
Abstract
Background Women with a history of preeclampsia (PE) have been shown to have up to five times the risk of developing later-life cardiovascular disease (CVD). While PE and CVD are known to share clinical and molecular characteristics, there are limited studies investigating their shared genomics (genetics, epigenetics or transcriptomics) variation over time. Therefore, we sought to systematically review the literature to identify longitudinal studies focused on the genomic progression to CVD following PE. Methods A literature search of primary sources through PubMed, Scopus, Web of Science and Embase via OVID was performed. Studies published from January 1, 1980, to July 28, 2023, that investigated genomics in PE and CVD were eligible for inclusion. Included studies were screened based on Cochrane systematic review guidelines in conjunction with the PRISMA 2020 checklist. Eligible articles were further assessed for quality using the Newcastle-Ottawa scale. Results A total of 9,231 articles were screened, with 14 studies subjected to quality assessment. Following further evaluation, six studies were included for the final review. All six of these studies were heterogeneous in regard to CVD/risk factor as outcome, gene mapping approach, and in different targeted genes. The associated genes were RGS2, LPA, and AQP3, alongside microRNAs miR-122-5p, miR-126-3p, miR-146a-5p, and miR-206. Additionally, 12 differentially methylated regions potentially linked to later-life CVD following PE were identified. The only common variable across all six studies was the use of a case-control study design. Conclusions Our results provide critical insight into the heterogeneous nature of genomic studies investigating CVD following PE and highlight the urgent need for longitudinal studies to further investigate the genetic variation underlying the progression to CVD following PE.
Collapse
Affiliation(s)
- Gayathry Krishnamurthy
- Menzies Institute for Medical Research, College of Health and Medicine, The University of Tasmania, Hobart, TAS, Australia
| | - Phuong Tram Nguyen
- Menzies Institute for Medical Research, College of Health and Medicine, The University of Tasmania, Hobart, TAS, Australia
| | - Bao Ngoc Tran
- Wicking Dementia Research and Education Center, College of Health and Medicine, The University of Tasmania, Hobart, TAS, Australia
| | - Hoang T. Phan
- Menzies Institute for Medical Research, College of Health and Medicine, The University of Tasmania, Hobart, TAS, Australia
| | - Shaun P. Brennecke
- Pregnancy Research Centre, Department of Maternal-Fetal Medicine, The Royal Women’s Hospital, Melbourne, VIC, Australia
- Department of Obstetrics and Gynaecology, The Royal Women’s Hospital, The University of Melbourne, Melbourne, VIC, Australia
| | - Eric K. Moses
- Menzies Institute for Medical Research, College of Health and Medicine, The University of Tasmania, Hobart, TAS, Australia
| | - Phillip E. Melton
- Menzies Institute for Medical Research, College of Health and Medicine, The University of Tasmania, Hobart, TAS, Australia
- School of Global and Population Health, The University of Western Australia, Crawley, WA, Australia
| |
Collapse
|
2
|
Matveyenko A, Seid H, Kim K, Ramakrishnan R, Thomas T, Matienzo N, Reyes-Soffer G. Association of free-living diet composition with plasma lipoprotein(a) levels in healthy adults. Lipids Health Dis 2023; 22:144. [PMID: 37670291 PMCID: PMC10478368 DOI: 10.1186/s12944-023-01884-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Accepted: 07/27/2023] [Indexed: 09/07/2023] Open
Abstract
BACKGROUND Lipoprotein (a) [Lp(a)] is an apoB100-containing lipoprotein with high levels being positively associated with atherosclerotic cardiovascular disease. Lp(a) levels are genetically determined. However, previous studies report a negative association between Lp(a) and saturated fatty acid intake. Currently, apoB100 lowering therapies are used to lower Lp(a) levels, and apheresis therapy is FDA approved for patients with extreme elevations of Lp(a). The current study analyzed the association of free-living diet components with plasma Lp(a) levels. METHODS Dietary composition data was collected during screening visits for enrollment in previously completed lipid and lipoprotein metabolism studies at Columbia University Irving Medical Center via a standardized protocol by registered dietitians using 24 hour recalls. Data were analyzed with the Nutrition Data System for Research (Version 2018). Diet quality was calculated using the Healthy Eating Index (HEI) score. Fasting plasma Lp(a) levels were measured via an isoform-independent ELISA and apo(a) isoforms were measured using gel electrophoresis. RESULTS We enrolled 28 subjects [Black (n = 18); Hispanic (n = 7); White (n = 3)]. The mean age was 48.3 ± 12.5 years with 17 males. Median level of Lp(a) was 79.9 nmol/L (34.4-146.0) and it was negatively associated with absolute (grams/day) and relative (percent of total calories) intake of dietary saturated fatty acids (SFA) (R = -0.43, P = 0.02, SFA …(% CAL): R = -0.38, P = 0.04), palmitic acid intake (R = -0.38, P = 0.05), and stearic acid intake (R = -0.40, P = 0.03). Analyses of associations with HEI score when stratified based on Lp(a) levels > or ≤ 100 nmol/L revealed no significant associations with any of the constituent factors. CONCLUSIONS Using 24 hour recall, we confirm previous findings that Lp(a) levels are negatively associated with dietary saturated fatty acid intake. Additionally, Lp(a) levels are not related to diet quality, as assessed by the HEI score. The mechanisms underlying the relationship of SFA with Lp(a) require further investigation.
Collapse
Affiliation(s)
- Anastasiya Matveyenko
- Department of Medicine, Columbia University Vagelos College of Physicians and Surgeons, New York, N.Y, USA
| | - Heather Seid
- Irving Institute for Clinical and Translational Research, Columbia University, New York, N.Y, USA
| | - Kyungyeon Kim
- Institute of Human Nutrition, Columbia University, New York, N.Y, USA
| | - Rajasekhar Ramakrishnan
- Center for Biomathematics, Department of Pediatrics, Columbia University Vagelos College of Physicians and Surgeons, New York, N.Y, USA
| | - Tiffany Thomas
- Department of Pathology and Cell Biology, Columbia University Vagelos College of Physicians and Surgeons, New York, N.Y, USA
| | - Nelsa Matienzo
- Department of Medicine, Columbia University Vagelos College of Physicians and Surgeons, New York, N.Y, USA
| | - Gissette Reyes-Soffer
- Department of Medicine, Columbia University Vagelos College of Physicians and Surgeons, New York, N.Y, USA.
| |
Collapse
|
3
|
Al Hageh C, Chacar S, Ghassibe-Sabbagh M, Platt DE, Henschel A, Hamdan H, Gauguier D, El Murr Y, Alefishat E, Chammas E, O’Sullivan S, Abchee A, Nader M, Zalloua PA. Elevated Lp(a) Levels Correlate with Severe and Multiple Coronary Artery Stenotic Lesions. Vasc Health Risk Manag 2023; 19:31-41. [PMID: 36703868 PMCID: PMC9871050 DOI: 10.2147/vhrm.s394134] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Accepted: 12/21/2022] [Indexed: 01/19/2023] Open
Abstract
Backgrounds and Aims The role of Lipoprotein(a) (Lp(a)) in increasing the risk of cardiovascular diseases is reported in several populations. The aim of this study is to investigate the correlation of high Lp(a) levels with the degree of coronary artery stenosis. Methods Two hundred and sixty-eight patients were enrolled for this study. Patients who underwent coronary artery angiography and who had Lp(a) measurements available were included in this study. Binomial logistic regressions were applied to investigate the association between Lp(a) and stenosis in the four major coronary arteries. The effect of LDL and HDL Cholesterol on modulating the association of Lp(a) with coronary artery disease (CAD) was also evaluated. Multinomial regression analysis was applied to assess the association of Lp(a) with the different degrees of stenosis in the four major coronary arteries. Results Our analyses showed that Lp(a) is a risk factor for CAD and this risk is significantly apparent in patients with HDL-cholesterol ≥35 mg/dL and in non-obese patients. A large proportion of the study patients with elevated Lp(a) levels had CAD even when exhibiting high HDL serum levels. Increased HDL with low Lp(a) serum levels were the least correlated with stenosis. A significantly higher levels of Lp(a) were found in patients with >50% stenosis in at least two major coronary vessels arguing for pronounced and multiple stenotic lesions. Finally, the derived variant (rs1084651) of the LPA gene was significantly associated with CAD. Conclusion Our study highlights the importance of Lp(a) levels as an independent biological marker of severe and multiple coronary artery stenosis.
Collapse
Affiliation(s)
- Cynthia Al Hageh
- Department of Molecular Biology and Genetics, College of Medicine and Health Sciences, Khalifa University of Science and Technology, Abu Dhabi, United Arab Emirates
| | - Stephanie Chacar
- Department of Physiology and Immunology, College of Medicine and Health Sciences, Khalifa University of Science and Technology, Abu Dhabi, United Arab Emirates
| | - Michella Ghassibe-Sabbagh
- Department of Natural Sciences, School of Arts and Sciences, Lebanese American University, Beirut, Lebanon
| | - Daniel E Platt
- Computational Biology Center, IBM TJ Watson Research Centre, Yorktown Hgts, NY, USA
| | - Andreas Henschel
- Department of Electrical Engineering and Computer, Khalifa University of Science and Technology, Abu Dhabi, United Arab Emirates
| | - Hamdan Hamdan
- Department of Physiology and Immunology, College of Medicine and Health Sciences, Khalifa University of Science and Technology, Abu Dhabi, United Arab Emirates
| | - Dominique Gauguier
- Université Paris Cité, INSERM UMR 1124, Paris, 75006, France,McGill University and Genome Quebec Innovation Centre, Montreal, QC, H3A 0G1, Canada
| | - Yara El Murr
- Department of Natural Sciences, School of Arts and Sciences, Lebanese American University, Beirut, Lebanon
| | - Eman Alefishat
- Department of Pharmacology, College of Medicine and Health Sciences, Khalifa University of Science and Technology, Abu Dhabi, United Arab Emirates,Center for Biotechnology, Khalifa University of Science and Technology, Abu Dhabi, 127788, United Arab Emirates
| | - Elie Chammas
- School of Medicine, Lebanese University, Beirut, Lebanon
| | - Siobhán O’Sullivan
- Department of Molecular Biology and Genetics, College of Medicine and Health Sciences, Khalifa University of Science and Technology, Abu Dhabi, United Arab Emirates
| | - Antoine Abchee
- Sheikh Shakhbout Medical City, Abu Dhabi, United Arab Emirates
| | - Moni Nader
- Department of Physiology and Immunology, College of Medicine and Health Sciences, Khalifa University of Science and Technology, Abu Dhabi, United Arab Emirates,Center for Biotechnology, Khalifa University of Science and Technology, Abu Dhabi, 127788, United Arab Emirates
| | - Pierre A Zalloua
- Department of Molecular Biology and Genetics, College of Medicine and Health Sciences, Khalifa University of Science and Technology, Abu Dhabi, United Arab Emirates,Center for Biotechnology, Khalifa University of Science and Technology, Abu Dhabi, 127788, United Arab Emirates,Harvard T.H. Chan School of Public Health, Boston, MA, USA,Correspondence: Pierre A Zalloua; Moni Nader, College of Medicine and Health Sciences, Khalifa University for Science and Technology, PO Box 127788, Abu Dhabi, United Arab Emirates, Email ;
| |
Collapse
|
4
|
A Novel Predictive Nomogram including Serum Lipoprotein a Level for Nonsentinel Lymph Node Metastases in Chinese Breast Cancer Patients with Positive Sentinel Lymph Node Metastases. DISEASE MARKERS 2021; 2021:7879508. [PMID: 34853623 PMCID: PMC8629655 DOI: 10.1155/2021/7879508] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Accepted: 10/26/2021] [Indexed: 11/18/2022]
Abstract
Background We developed a new nomogram combining serum biomarkers with clinicopathological features to improve the accuracy of prediction of nonsentinel lymph node (SLN) metastases in Chinese breast cancer patients. Methods We enrolled 209 patients with breast cancer who underwent SLN biopsy and axillary lymph node dissection. We evaluated the relationships between non-SLN metastases and clinicopathologic features, as well as preoperative routine tests of blood indexes, tumor markers, and serum lipids, including lipoprotein a (Lp(a)). Risk factors for non-SLN metastases were identified by logistic regression analysis. The nomogram was created using the R program to predict the risk of non-SLN metastases in the training set. Receiver operating characteristic (ROC) analysis was applied to assess the predictive value of the nomogram model in the validation set. Results Lp(a) was significantly associated with non-SLN metastasis status. Compared with the MSKCC model, the predictive ability of our new nomogram that combined Lp(a) level and clinical variables (pathologic tumor size, lymphovascular invasion, multifocality, and positive/negative SLN numbers) was significantly greater (AUC: 0.732, 95% CI: 0.643–0.821) (C-index: 0.703, 95% CI: 0.656–0.791) in the training cohorts and also performed well in the validation cohorts (C-index: 0.773, 95% CI: 0.681–0.865). Moreover, the new nomogram with Lp(a) improved the accuracy (12.10%) of identification of patients with non-SLN metastases (NRI: 0.121; 95% CI: 0.081–0.202; P = 0.011). Conclusions This novel nomogram based on preoperative serum indexes combined with clinicopathologic features facilitates accurate prediction of risk of non-SLN metastases in Chinese patients with breast cancer.
Collapse
|
5
|
Gu Y, Gan S, Bian S, Meng G, Zhang Q, Liu L, Wu H, Yao Z, Zhang S, Wang Y, Zhang T, Wang X, Cao X, Li H, Liu Y, Li X, Wang X, Wang X, Sun S, Zhou M, Jia Q, Song K, Wu Y, Wu XH, Niu K. The association between daily yogurt consumption and serum lipid profiles in the general adult population: the TCLSIH cohort study. Int J Food Sci Nutr 2021; 73:415-423. [PMID: 34749580 DOI: 10.1080/09637486.2021.1993155] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
The purpose of this cross-sectional study is to determine the association between yogurt consumption and lipid profiles in the general Chinese population. In this cross-sectional study, a total of 26,824 participants were included from Tianjin, China. Lipid profiles were determined by automated biochemical analyser. Yogurt consumption frequency was assessed by a validated food frequency questionnaire. Analysis of covariance was used to determine the association between daily yogurt consumption and lipid profiles. In the final multivariate model, daily yogurt consumption was negatively associated with triglyceride (TG) (p for trend <0.001) and positively associated with high density lipoprotein cholesterol (HDL-C) (p for trend = 0.02). There were no significant differences (p for trend >0.05) between daily yogurt consumption and total cholesterol (TC) or low density lipoprotein cholesterol (LDL-C). Results suggested that higher daily yogurt consumption was negatively correlated with TG and positively correlated with HDL-C in the general Chinese population.
Collapse
Affiliation(s)
- Yeqing Gu
- Nutrition and Radiation Epidemiology Research Center, Institute of Radiation Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
| | - Shinan Gan
- Nutritional Epidemiology Institute and School of Public Health, Tianjin Medical University, Tianjin, China
| | - Shanshan Bian
- Nutritional Epidemiology Institute and School of Public Health, Tianjin Medical University, Tianjin, China
| | - Ge Meng
- Nutritional Epidemiology Institute and School of Public Health, Tianjin Medical University, Tianjin, China.,Department of Toxicology and Sanitary Chemistry, School of Public Health, Tianjin Medical University, Tianjin, China
| | - Qing Zhang
- Health Management Centre, Tianjin Medical University General Hospital, Tianjin, China
| | - Li Liu
- Health Management Centre, Tianjin Medical University General Hospital, Tianjin, China
| | - Hongmei Wu
- Nutritional Epidemiology Institute and School of Public Health, Tianjin Medical University, Tianjin, China
| | - Zhanxin Yao
- Tianjin Institute of Health and Environmental Medicine, Tianjin, China
| | - Shunming Zhang
- Nutritional Epidemiology Institute and School of Public Health, Tianjin Medical University, Tianjin, China
| | - Yawen Wang
- Nutritional Epidemiology Institute and School of Public Health, Tianjin Medical University, Tianjin, China
| | - Tingjing Zhang
- Nutritional Epidemiology Institute and School of Public Health, Tianjin Medical University, Tianjin, China
| | - Xuena Wang
- Nutritional Epidemiology Institute and School of Public Health, Tianjin Medical University, Tianjin, China
| | - Xingqi Cao
- Nutritional Epidemiology Institute and School of Public Health, Tianjin Medical University, Tianjin, China
| | - Huiping Li
- Nutritional Epidemiology Institute and School of Public Health, Tianjin Medical University, Tianjin, China
| | - Yunyun Liu
- Nutritional Epidemiology Institute and School of Public Health, Tianjin Medical University, Tianjin, China
| | - Xiaoyue Li
- Nutritional Epidemiology Institute and School of Public Health, Tianjin Medical University, Tianjin, China
| | - Xiaohe Wang
- Nutritional Epidemiology Institute and School of Public Health, Tianjin Medical University, Tianjin, China
| | - Xing Wang
- Health Management Centre, Tianjin Medical University General Hospital, Tianjin, China
| | - Shaomei Sun
- Health Management Centre, Tianjin Medical University General Hospital, Tianjin, China
| | - Ming Zhou
- Health Management Centre, Tianjin Medical University General Hospital, Tianjin, China
| | - Qiyu Jia
- Health Management Centre, Tianjin Medical University General Hospital, Tianjin, China
| | - Kun Song
- Health Management Centre, Tianjin Medical University General Hospital, Tianjin, China
| | - Yuntang Wu
- Nutritional Epidemiology Institute and School of Public Health, Tianjin Medical University, Tianjin, China
| | - Xiao-Hui Wu
- College of Pharmacy, Tianjin Medical University, Tianjin, China
| | - Kaijun Niu
- Nutrition and Radiation Epidemiology Research Center, Institute of Radiation Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China.,Nutritional Epidemiology Institute and School of Public Health, Tianjin Medical University, Tianjin, China.,Health Management Centre, Tianjin Medical University General Hospital, Tianjin, China.,Tianjin Key Laboratory of Environment, Nutrition and Public Health, Tianjin, China.,Center for International Collaborative Research on Environment, Nutrition and Public Health, Tianjin, China
| |
Collapse
|
6
|
Satterfield BA, Dikilitas O, Safarova MS, Clarke SL, Tcheandjieu C, Zhu X, Bastarache L, Larson EB, Justice AE, Shang N, Rosenthal EA, Shah AS, Namjou-Khales B, Urbina EM, Wei WQ, Feng Q, Jarvik GP, Hebbring SJ, de Andrade M, Manolio TA, Assimes TL, Kullo IJ. Associations of Genetically Predicted Lp(a) (Lipoprotein [a]) Levels With Cardiovascular Traits in Individuals of European and African Ancestry. CIRCULATION. GENOMIC AND PRECISION MEDICINE 2021; 14:e003354. [PMID: 34282949 PMCID: PMC8634549 DOI: 10.1161/circgen.120.003354] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
BACKGROUND Lp(a) (lipoprotein [a]) levels are higher in individuals of African ancestry (AA) than in individuals of European ancestry (EA). We examined associations of genetically predicted Lp(a) levels with (1) atherosclerotic cardiovascular disease subtypes: coronary heart disease, cerebrovascular disease, peripheral artery disease, and abdominal aortic aneurysm and (2) nonatherosclerotic cardiovascular disease phenotypes, stratified by ancestry. METHODS We performed (1) Mendelian randomization analyses for previously reported cardiovascular associations and (2) Mendelian randomization-phenome-wide association analyses for novel associations. Analyses were stratified by ancestry in electronic Medical Records and Genomics, United Kingdom Biobank, and Million Veteran Program cohorts separately and in a combined cohort of 804 507 EA and 103 580 AA participants. RESULTS In Mendelian randomization analyses using the combined cohort, a 1-SD genetic increase in Lp(a) level was associated with atherosclerotic cardiovascular disease subtypes in EA-odds ratio and 95% CI for coronary heart disease 1.28 (1.16-1.41); cerebrovascular disease 1.14 (1.07-1.21); peripheral artery disease 1.22 (1.11-1.34); abdominal aortic aneurysm 1.28 (1.17-1.40); in AA, the effect estimate was lower than in EA and nonsignificant for coronary heart disease 1.11 (0.99-1.24) and cerebrovascular disease 1.06 (0.99-1.14) but similar for peripheral artery disease 1.16 (1.01-1.33) and abdominal aortic aneurysm 1.34 (1.11-1.62). In EA, a 1-SD genetic increase in Lp(a) level was associated with aortic valve disorders 1.34 (1.10-1.62), mitral valve disorders 1.18 (1.09-1.27), congestive heart failure 1.12 (1.05-1.19), and chronic kidney disease 1.07 (1.01-1.14). In AA, no significant associations were noted for aortic valve disorders 1.08 (0.94-1.25), mitral valve disorders 1.02 (0.89-1.16), congestive heart failure 1.02 (0.95-1.10), or chronic kidney disease 1.05 (0.99-1.12). Mendelian randomization-phenome-wide association analyses identified novel associations in EA with arterial thromboembolic disease, nonaortic aneurysmal disease, atrial fibrillation, cardiac conduction disorders, and hypertension. CONCLUSIONS Many cardiovascular associations of genetically increased Lp(a) that were significant in EA were not significant in AA. Lp(a) was associated with atherosclerotic cardiovascular disease in four major arterial beds in EA but only with peripheral artery disease and abdominal aortic aneurysm in AA. Additionally, novel cardiovascular associations were detected in EA.
Collapse
Affiliation(s)
| | - Ozan Dikilitas
- Dept of Cardiovascular Medicine, Mayo Clinic, Rochester, MN
| | | | - Shoa L. Clarke
- VA Palo Alto Health Care System, Palo Alto
- Division of Cardiovascular Medicine, Dept of Medicine, Stanford Univ School of Medicine, Stanford, CA
| | - Catherine Tcheandjieu
- VA Palo Alto Health Care System, Palo Alto
- Division of Cardiovascular Medicine, Dept of Medicine, Stanford Univ School of Medicine, Stanford, CA
- Dept of Pediatric Cardiology, Stanford Univ School of Medicine, Stanford, CA
| | - Xiang Zhu
- VA Palo Alto Health Care System, Palo Alto
- Dept of Statistics, The Pennsylvania State Univ, University Park, PA
- Huck Institutes of the Life Sciences, The Pennsylvania State Univ, University Park, PA
- Dept of Statistics, Stanford Univ, Stanford, CA
| | - Lisa Bastarache
- Dept of Biomedical Informatics, Vanderbilt Univ, Nashville, TN
| | - Eric B. Larson
- Kaiser Permanente Washington Health Research Institutes, Seattle, WA
| | | | - Ning Shang
- Dept of Biomedical Informatics, Columbia Univ, New York, NY
| | | | - Amy Sanghavi Shah
- Division of Endocrinology, Cincinnati Children’s Hospital Medical Center & Univ of Cincinnati
| | - Bahram Namjou-Khales
- Center for Autoimmune Genomics and Etiology, Cincinnati Children’s Hospital Medical Center & Dept of Pediatrics, Univ of Cincinnati, College of Medicine, Cincinnati, OH
| | - Elaine M. Urbina
- Division of Endocrinology, Heart Institute, Cincinnati Children’s Hospital Medical Center & Univ of Cincinnati
| | - Wei-Qi Wei
- Dept of Biomedical Informatics, Vanderbilt Univ, Nashville, TN
| | - QiPing Feng
- Division of Clinical Pharmacology, Dept of Medicine, Vanderbilt Univ Medical Center, Nashville, TN
| | - Gail P. Jarvik
- Division of Medical Genetics, Dept of Medicine, Univ of Washington, Seattle, WA
| | - Scott J. Hebbring
- Center for Precision Medicine, Marshfield Clinic Research Institute, WI
| | - Mariza de Andrade
- Dept of Cardiovascular Medicine, Dept of Health Sciences Research, Mayo Clinic, Rochester, MN
| | - Teri A. Manolio
- Division of Genomic Medicine, National Human Genome Research Institute, Bethesda, MD
| | | | - Iftikhar J. Kullo
- Dept of Cardiovascular Medicine, Mayo Clinic, Rochester, MN
- Dept of Cardiovascular Medicine, Gonda Vascular Center, Mayo Clinic, Rochester, MN
| |
Collapse
|
7
|
Babaahmadi-Rezaei H, Kheirollah A, Hesam S, Ayashi S, Aberumand M, Adel MH, Zamanpour M, Alasvand M, Amozgari Z, Noor-Behbahani M, Niknam Z. Decreased lipoprotein (a) and serum high-sensitivity C-reactive protein levels in male patients with atherosclerosis after supplementation with ginger: A randomized controlled trial. ARYA ATHEROSCLEROSIS 2021; 16:153-160. [PMID: 33598035 PMCID: PMC7867307 DOI: 10.22122/arya.v16i4.2011] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
BACKGROUND Although the antioxidant properties of ginger have been revealed, there is little available information on the effectiveness of ginger on inflammatory disorders such as atherosclerosis. This study was carried out to examine the effect of ginger on improving the complication of atherosclerosis. METHODS This study was a double-blind, placebo-controlled, randomized clinical trial conducted on patients with atherosclerosis. Participants in the ginger and control groups received 1600 mg of powdered ginger or placebo (wheat flour) in capsules daily for 8 weeks. Weight, body mass index (BMI), fasting blood sugar (FBS), cholesterol, triglyceride (TG), low-density lipoprotein (LDL), very-low-density lipoprotein (VLDL), high-density lipoprotein (HDL), lipoprotein (a) [Lp(a)], high-sensitivity C-reactive protein (hs-CRP), and total anti-oxidant capacity (TAC) were assessed before and after the intervention. RESULTS Ginger consumption in the intervention group significantly reduced serum Lp(a) level (27.25 ± 1.30 ng/ml vs. 23.57 ± 0.97 ng/ml) (P = 0.040) and also the level of hs-CRP in the intervention group was 1.90 ± 0.33 µg/ml and 1.24 ± 0.15 µg/ml (P = 0.010) before and after intervention, respectively, but the levels of Lp(a) and hs-CRP were not decreased significantly in the placebo group. The level of TAC in the ginger group was 0.71 ± 0.05 mM and after the trial was 0.57 ± 0.04 mM (P = 0.050); no significant differences were seen in TAC when ginger was administered at 1600 mg/daily for 60 days. Also the level of Lp(a) and hs-CRP but not TAC reduced significantly in ginger group compared to placebo group after intervention. CONCLUSION This study showed that ginger had anti-atherosclerosis and anti-glycemic properties associated through a significant decreased Lp(a) and FBS in patients with atherosclerosis supplemented with ginger.
Collapse
Affiliation(s)
- Hossein Babaahmadi-Rezaei
- Associate Professor, Cellular and Molecular Research Center AND Department of Clinical Biochemistry, School of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Alireza Kheirollah
- Associate Professor, Cellular and Molecular Research Center AND Department of Clinical Biochemistry, School of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Saeed Hesam
- Assistant Professor, Department of Biostatistics and Epidemiology, School of Public Health, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Saleh Ayashi
- PhD Candidate, Cellular and Molecular Research Center AND Department of Clinical Biochemistry, School of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Mohammad Aberumand
- Associate Professor, Cellular and Molecular Research Center AND Department of Clinical Biochemistry, School of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Mohammad Hassan Adel
- Associate Professor, Atherosclerosis Research Center AND Department of Cardiology, School of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Masoumeh Zamanpour
- Cellular and Molecular Research Center AND Department of Clinical Biochemistry, School of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Maryam Alasvand
- Assistant Professor, Atherosclerosis Research Center AND Department of Cardiology, School of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Zohreh Amozgari
- Lecturer, Cellular and Molecular Research Center AND Department of Clinical Biochemistry, School of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Mojgan Noor-Behbahani
- Cellular and Molecular Research Center AND Department of Clinical Biochemistry, School of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Zahra Niknam
- Cellular and Molecular Research Center AND Department of Clinical Biochemistry, School of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| |
Collapse
|
8
|
Liu T, Yoon WS, Lee SR. Recent Updates of Lipoprotein(a) and Cardiovascular Disease. Chonnam Med J 2021; 57:36-43. [PMID: 33537217 PMCID: PMC7840349 DOI: 10.4068/cmj.2021.57.1.36] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Revised: 12/14/2020] [Accepted: 12/16/2020] [Indexed: 12/22/2022] Open
Abstract
In recent years, epidemiological studies, genome-wide association studies, and Mendelian randomization studies have shown a strong association between increased levels of lipoproteins and increased risks of coronary heart disease and cardiovascular disease (CVD). Although lipoprotein(a) [Lp(a)] was an independent risk factor for ASCVD, the latest international clinical guidelines do not recommend direct reduction of plasma Lp(a) concentrations. The main reason was that there is no effective clinical medicine that directly lowers plasma Lp(a) concentrations. However, recent clinical trials have shown that proprotein convertase subtilisin/kexin-type 9 inhibitors (PCSK9) and second-generation antisense oligonucleotides can effectively reduce plasma Lp(a) levels. This review will present the structure, pathogenicity, prognostic evidences, and recent advances in therapeutic drugs for Lp(a).
Collapse
Affiliation(s)
- Taili Liu
- Division of Cardiology, Department of Internal Medicine, Chonbuk National University Hospital, Jeonju, Korea
| | - Won-Sik Yoon
- Division of Cardiology, Department of Internal Medicine, Chonbuk National University Hospital, Jeonju, Korea
| | - Sang-Rok Lee
- Division of Cardiology, Department of Internal Medicine, Chonbuk National University Hospital, Jeonju, Korea
| |
Collapse
|
9
|
Li H, Pan K, Meng Y, Deng J, Zhang P, Song W, Li S. Mutual promotions between periodontitis and vascular calcification by rat animal model. J Periodontal Res 2020; 55:810-820. [PMID: 32996601 DOI: 10.1111/jre.12757] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2019] [Revised: 03/09/2020] [Accepted: 04/06/2020] [Indexed: 01/07/2023]
Abstract
OBJECTIVE AND BACKGROUND To study the relationship between periodontitis and vascular calcification by establishing rat model of chronic periodontitis and vascular calcification. METHODS Forty male Wistar rats were divided into four groups randomly: control group, periodontitis group, vascular calcification group, and compound periodontitis and calcification group. Each group rats accepted the corresponding manages to establish the animal model. Clinical examinations and hematoxylin and eosin staining of periodontal tissue were taken to test the periodontal model; calcium assay, alkaline phosphatase activity, expression of mineral-related factors including osteopontin, alkaline phosphatase, core-binding factor-α1 and bone sialoprotein, hematoxylin and eosin staining and von Kossa staining of vascular tissue were taken to test the vascular calcification model; inflammatory factors including C-reactive protein, interleukin-1β, tumor necrosis factor-α, interleukin-6, prostaglandin E2, and serum lipid in serum were also detected at the same time. RESULTS The rat model was established. Inflammation of periodontal tissue and alveolar bone resorption in compound group and periodontitis group were more obvious than those in control group and vascular calcification group (P < .05). However, the calcium assay, alkaline phosphatase activity, and mineralized deposition in vascular calcification group and compound group were higher than those in control group and periodontitis group (P < .05), and compound group were the highest (P < .05); as for serum lipid, the level of total cholesterol and low-density lipoprotein-cholesterol in compound group and vascular calcification group were higher than that in control group and periodontitis group (P < .05), and compound group was the highest (P <.05); but the level of high-density lipoprotein cholesterol was higher in control group and periodontitis group. Inflammatory factors expression in serum were higher in compound group and periodontitis group, while mineral-related factors expression were higher in compoundgroup and vascular calcification group. CONCLUSION There are some mutual promotions between periodontitis and vascular calcification, which might be related to the increasing inflammatory factors, lipids level, and mineral-related factors.
Collapse
Affiliation(s)
- Huixu Li
- The Affiliated Hospital of Qingdao University, Qingdao, China.,School of Stomatology of Qingdao University, Qingdao, China.,Department of Endodontics, Tianjin Stomatological Hospital, Hospital of Stomatology, Nankai University, TianJin, China
| | - Keqing Pan
- The Affiliated Hospital of Qingdao University, Qingdao, China.,School of Stomatology of Qingdao University, Qingdao, China
| | - Yun Meng
- The Affiliated Hospital of Qingdao University, Qingdao, China.,School of Stomatology of Qingdao University, Qingdao, China.,Department of Stomatology, The Traditional Chinese Medicine Hospital of Tianjin Dongli District, Tianjin, China
| | - Jing Deng
- The Affiliated Hospital of Qingdao University, Qingdao, China.,School of Stomatology of Qingdao University, Qingdao, China
| | - Pengmei Zhang
- The Affiliated Hospital of Qingdao University, Qingdao, China.,School of Stomatology of Qingdao University, Qingdao, China.,Department of Stomatology, The Huikang Hospital, Qingdao, China
| | - Wenbin Song
- The Affiliated Hospital of Qingdao University, Qingdao, China.,School of Stomatology of Qingdao University, Qingdao, China
| | - Shu Li
- Department of Periodontology, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration, Jinan, China
| |
Collapse
|
10
|
Hamamura H, Adachi H, Enomoto M, Fukami A, Nakamura S, Nohara Y, Morikawa N, Sakaue A, Toyomasu K, Yamamoto M, Fukumoto Y. Serum Proprotein Convertase Subtilisin/Kexin Type 9 (PCSK9) is Independently Associated with Insulin Resistance, Triglycerides, Lipoprotein(a) Levels but not Low-Density Lipoprotein Cholesterol Levels in a General Population. J Atheroscler Thromb 2020; 28:329-337. [PMID: 32624555 PMCID: PMC8147011 DOI: 10.5551/jat.56390] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Aim: Proprotein convertase subtilisin/kexin type 9 (PCSK9) has been identified as an important regulator of low-density lipoprotein (LDL) receptor processing. Evolocumab and alirocumab are PCSK9 inhibitors; however, little is known about the association between PCSK9 levels and lipid profiles in a general population. Because PCSK9 inhibitors have LDL-C lowering effects, we investigated whether there is a positive correlation between serum PCSK9 levels and LDL-C or lipoprotein(a) [Lp(a)]. Methods: In Uku town, 674 residents (mean age; 69.2 ± 8.3 years) received health check-ups. The participants underwent a physical examination and blood tests, including PCSK9 and Lp(a). Serum PCSK9 and Lp(a) were measured by ELISA and Latex methods, respectively. HOMA-IR was calculated by fasting plasma glucose × insulin levels/405. Results: The mean (range) of PCSK9 and Lp(a) were 211.2 (49–601) ng/mL and 60 (1–107) mg/dL, respectively. Because of a skewed distribution, the log-transformed values were used. With univariate linear regression analysis, PCSK9 levels were associated with Lp(a) (p = 0.028), triglycerides (p < 0.001), and HOMA-IR (p < 0.001), but not with LDL-C (p = 0.138) levels. Multiple stepwise regression analysis revealed that serum PCSK9 levels were independently associated with triglycerides (p < 0.001), Lp(a) (p = 0.033) and HOMA-IR (p = 0.041). Conclusions: PCSK-9 is independently associated with triglycerides, Lp(a) levels, and HOMA-IR, but not LDL-C, in a relatively large general population sample.
Collapse
Affiliation(s)
- Hitoshi Hamamura
- Department of Internal Medicine, Division of Cardio-Vascular Medicine, Kurume University School of Medicine
| | - Hisashi Adachi
- Department of Internal Medicine, Division of Cardio-Vascular Medicine, Kurume University School of Medicine.,Department of Community Medicine, Kurume University School of Medicine
| | - Mika Enomoto
- Department of Internal Medicine, Division of Cardio-Vascular Medicine, Kurume University School of Medicine
| | - Ako Fukami
- Department of Internal Medicine, Division of Cardio-Vascular Medicine, Kurume University School of Medicine
| | - Sachiko Nakamura
- Department of Internal Medicine, Division of Cardio-Vascular Medicine, Kurume University School of Medicine
| | - Yume Nohara
- Department of Internal Medicine, Division of Cardio-Vascular Medicine, Kurume University School of Medicine
| | - Nagisa Morikawa
- Department of Internal Medicine, Division of Cardio-Vascular Medicine, Kurume University School of Medicine
| | - Akiko Sakaue
- Department of Internal Medicine, Division of Cardio-Vascular Medicine, Kurume University School of Medicine
| | - Kenta Toyomasu
- Department of Internal Medicine, Division of Cardio-Vascular Medicine, Kurume University School of Medicine
| | - Maki Yamamoto
- Department of Internal Medicine, Division of Cardio-Vascular Medicine, Kurume University School of Medicine
| | - Yoshihiro Fukumoto
- Department of Internal Medicine, Division of Cardio-Vascular Medicine, Kurume University School of Medicine
| |
Collapse
|
11
|
Lipoprotein(a) and long-term recurrent infarction after an episode of ST-segment elevation acute myocardial infarction. Coron Artery Dis 2020; 31:378-384. [PMID: 32040026 DOI: 10.1097/mca.0000000000000852] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
BACKGROUND In established ischemic heart disease, the relationship between lipoprotein(a) and new cardiovascular events showed contradictory results. Our aim was to assess the relationship between lipoprotein(a) and very long-term recurrent myocardial infarction (MI) after an index episode of ST-segment elevation acute myocardial infarction (STEMI). METHODS We included 435 consecutive STEMI patients discharged from October 2000 to June 2003 in a single teaching center. The relationship between lipoprotein(a) at discharge and recurrent MI was evaluated through negative binomial regression and Cox regression analysis. RESULTS The mean age was 65 years (55-74 years), 25.5% were women, 34.7% were diabetic, and 66% had a MI of anterior location. Fibrinolysis, rescue, or primary angioplasty was performed in 215 (49.4%), 19 (4.4%), and 18 (4.1%) patients, respectively. The median lipoprotein(a) was 30.4 mg/dL (12-59.4 mg/dL). After a median follow-up of 9.6 years (4.1-15 years), 180 (41.4%) deaths and 187 MI in 133 (30.6%) patients were recorded. After a multivariate adjustment, the risk gradient of lipoprotein(a) showed a neutral effect along most of the continuum and only extreme higher values identified those at higher risk of recurrent MI (P = 0.020). Those with lipoprotein(a) values >95th percentile (≥135 mg/dL) showed a higher risk of recurrent MI (incidence rate ratio, 2.34; 95% confidence interval, 1.37-4.02; P = 0.002). Lipoprotein(a) was not related to the risk of mortality (P = 0.245). CONCLUSIONS After an episode of STEMI, only extreme high values of lipoprotein(a) were associated with an increased risk of long-term recurrent MI.
Collapse
|
12
|
Jawi MM, Frohlich J, Chan SY. Lipoprotein(a) the Insurgent: A New Insight into the Structure, Function, Metabolism, Pathogenicity, and Medications Affecting Lipoprotein(a) Molecule. J Lipids 2020; 2020:3491764. [PMID: 32099678 PMCID: PMC7016456 DOI: 10.1155/2020/3491764] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2019] [Accepted: 08/17/2019] [Indexed: 12/15/2022] Open
Abstract
Lipoprotein(a) [Lp(a)], aka "Lp little a", was discovered in the 1960s in the lab of the Norwegian physician Kåre Berg. Since then, we have greatly improved our knowledge of lipids and cardiovascular disease (CVD). Lp(a) is an enigmatic class of lipoprotein that is exclusively formed in the liver and comprises two main components, a single copy of apolipoprotein (apo) B-100 (apo-B100) tethered to a single copy of a protein denoted as apolipoprotein(a) apo(a). Plasma levels of Lp(a) increase soon after birth to a steady concentration within a few months of life. In adults, Lp(a) levels range widely from <2 to 2500 mg/L. Evidence that elevated Lp(a) levels >300 mg/L contribute to CVD is significant. The improvement of isoform-independent assays, together with the insight from epidemiologic studies, meta-analyses, genome-wide association studies, and Mendelian randomization studies, has established Lp(a) as the single most common independent genetically inherited causal risk factor for CVD. This breakthrough elevated Lp(a) from a biomarker of atherosclerotic risk to a target of therapy. With the emergence of promising second-generation antisense therapy, we hope that we can answer the question of whether Lp(a) is ready for prime-time clinic use. In this review, we present an update on the metabolism, pathophysiology, and current/future medical interventions for high levels of Lp(a).
Collapse
Affiliation(s)
- Motasim M. Jawi
- Healthy Heart Program, St. Paul's Hospital, Vancouver V6Z 1Y6, Canada
- Division of Experimental Medicine, Department of Medicine, Faculty of Medicine, University of British Columbia, Vancouver V5Z 1M9, Canada
- Department of Clinical PhysiologyCorrection: Department of Physiology, University of Jeddah, P.O. Box: 24, Jeddah 21959, Saudi Arabia
| | - Jiri Frohlich
- Healthy Heart Program, St. Paul's Hospital, Vancouver V6Z 1Y6, Canada
- Department of Pathology and Laboratory Medicine, Faculty of Medicine, University of British Columbia, Vancouver, British Columbia V6T 2B5, Canada
| | - Sammy Y. Chan
- Healthy Heart Program, St. Paul's Hospital, Vancouver V6Z 1Y6, Canada
- Department of Medicine, Division of Cardiology, University of British Columbia, Vancouver V5Z 1M9, Canada
| |
Collapse
|
13
|
Wang L, Cong H, Zhang J, Hu Y, Wei A, Zhang Y. Prognostic Value of Lipoprotein(a) Levels in Patients Undergoing Coronary Angiography for Premature Acute Coronary Syndromes. Angiology 2019; 71:160-166. [PMID: 31722547 DOI: 10.1177/0003319719886493] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Little is known about the association between lipoprotein(a) [Lp(a)] levels and future ischemic cardiovascular events in patients with premature acute coronary syndrome (ACS). A total of 1464 consecutive patients who underwent coronary angiography for premature ACS (males <45 years and females <55 years) were enrolled in this study. Patients were divided into quartiles according to serum Lp(a) levels (Q1: ≤11.1 nmol/L; Q2: 11.1-27.7 nmol/L; Q3: 27.7-79.3 nmol/L; and Q4: >79.3 nmol/L). Major adverse cardiovascular events (MACEs) increased with Lp(a) quartiles after 2-year follow-up (among quartiles, respectively; P = .001). Kaplan-Meier curves revealed significant differences in event-free survival rates among Lp(a) quartile groups ( P = .001). Multivariate Cox proportional hazards regression analysis indicated that serum Lp(a) level was an independent predictor of MACE either as a continuous variable (hazard ratio [HR]: 1.002, 95% confidence interval [CI]: 1.001-1.004; P = .009) or as a categorical variable (HR: 1.443, 95% CI: 1.074-1.937; P = .015). Furthermore, Lp(a) levels (as a variable) significantly improved the prognostic value for MACE. These findings suggest that Lp(a) measurement has value for cardiovascular risk stratification in patients with premature ACS.
Collapse
Affiliation(s)
- Le Wang
- Department of Cardiology, Tianjin Chest Hospital, Tianjin, China
| | - Hongliang Cong
- Department of Cardiology, Tianjin Chest Hospital, Tianjin, China
| | - Jingxia Zhang
- Department of Cardiology, Tianjin Chest Hospital, Tianjin, China
| | - Yuecheng Hu
- Department of Cardiology, Tianjin Chest Hospital, Tianjin, China
| | - Ao Wei
- Department of Cardiology, Tianjin Chest Hospital, Tianjin, China
| | - Yingyi Zhang
- Department of Cardiology, Tianjin Chest Hospital, Tianjin, China
| |
Collapse
|
14
|
Pavanello C, Pirazzi C, Bjorkman K, Sandstedt J, Tarlarini C, Mosca L, Romeo S, Calabresi L, Mancina RM. Individuals with familial hypercholesterolemia and cardiovascular events have higher circulating Lp(a) levels. J Clin Lipidol 2019; 13:778-787.e6. [DOI: 10.1016/j.jacl.2019.06.011] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2018] [Revised: 06/24/2019] [Accepted: 06/27/2019] [Indexed: 10/26/2022]
|
15
|
Stulnig TM, Morozzi C, Reindl-Schwaighofer R, Stefanutti C. Looking at Lp(a) and Related Cardiovascular Risk: from Scientific Evidence and Clinical Practice. Curr Atheroscler Rep 2019; 21:37. [PMID: 31350625 DOI: 10.1007/s11883-019-0803-9] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
PURPOSE OF REVIEW A considerable body of data from genetic and epidemiological studies strongly support a causal relationship between high lipoprotein(a) [Lp(a)] levels, and the development of atherosclerosis and cardiovascular disease. This relationship is continuous, unrelated to Lp(a) threshold, and independent of low-density lipoprotein (LDL) and high-density lipoprotein (HDL) cholesterol levels. Unfortunately, the mechanism(s) through which Lp(a) promotes atherosclerosis are not clarified yet. Suggested hypotheses include: an increased Lp(a)-associated cholesterol entrapment in the arterial intima followed by inflammatory cell recruitment, abnormal upload of proinflammatory oxidized phospholipids, impaired fibrinolysis by inhibition of plasminogen activation, and enhanced coagulation, through inhibition of the tissue factor pathway inhibitor. This review is aimed at summarizing the available evidence on the topic. RECENT FINDINGS There are two clinical forms, isolated hyperlipidemia(a) [HyperLp(a)] with acceptable LDL-C levels (< 70 mg/dL), and combined elevation of Lp(a) and LDL-C in plasma. To date, no drugs that selectively decrease Lp(a) are available. Some novel lipid-lowering drugs can lower Lp(a) levels, but to a limited extent, as their main effect is aimed at decreasing LDL-C levels. Significant Lp(a) lowering effects were obtained with nicotinic acid at high doses. However, adverse effects apart, nicotinic acid is no longer prescribed and available in Europe for clinical use, after European Agency of Medicines (EMA) ban. The only effective therapeutic option for now is Lipoprotein Apheresis (LA), albeit with some limitations. Lastly, it is to be acknowledged that the body of evidence confirming that reducing plasma isolated elevation of Lp(a) brings cardiovascular benefit is still insufficient. However, the growing bulk of clinical, genetic, mechanistic, and epidemiological available evidence strongly suggests that Lp(a) is likely to be the smoking gun.
Collapse
Affiliation(s)
- Thomas M Stulnig
- Clinical Division of Endocrinology and Metabolism - Department of Medicine III - Medical University of Vienna, Universitätsring 1, 1010, Wien, Austria
| | - Claudia Morozzi
- Department of Molecular Medicine, Lipid Clinic and Atherosclerosis Prevention Centre -"Sapienza" University of Rome, Extracorporeal Therapeutic Techniques Unit, Regional Centre for Rare Diseases, Immunohematology and Transfusion Medicine, "Umberto I" Hospital, Rome, Italy
| | - Roman Reindl-Schwaighofer
- Division of Nephrology and Dialysis, Department of Medicine III, Medical University of Vienna, Spitalgasse 23, 1090, Wien, Austria
| | - Claudia Stefanutti
- Department of Molecular Medicine, Lipid Clinic and Atherosclerosis Prevention Centre -"Sapienza" University of Rome, Extracorporeal Therapeutic Techniques Unit, Regional Centre for Rare Diseases, Immunohematology and Transfusion Medicine, "Umberto I" Hospital, Rome, Italy.
| |
Collapse
|
16
|
Vadini F, Santilli F, Casalini G, dell'Isola M, Iuliani O, D'Ardes D, Lattanzio L, Di Nicola M, Di Iorio G, Accorsi P. Homocysteine and education but not lipoprotein (a) predict estimated 10-year risk of cardiovascular disease in blood donors: a community based cross-sectional study. BMC Cardiovasc Disord 2019; 19:177. [PMID: 31349819 PMCID: PMC6660663 DOI: 10.1186/s12872-019-1157-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2019] [Accepted: 07/15/2019] [Indexed: 11/24/2022] Open
Abstract
Background With aging of the population, screening and prevention health programs for blood donors will increasingly be a priority. We aimed at: assessing the 10 year-cardiovascular disease (CVD) risk in blood donors, according to Italian CUORE risk score (CRS); determining the association of homocysteine (Hcy), lipoprotein (Lp)(a) and socio-demographic or lifestyle variables with estimated 10-year CVD risk. Methods Between June 2015 and July 2017, 1,447 (61.2% men) unselected blood donors (aged 18–69 years) were enrolled at the Blood Transfusion Service of the Pescara General Hospital, Italy. The project entailed evaluation of unalterable (age and gender) and modifiable CV risk factors (total cholesterol, HDL, LDL, triglycerides, fasting glucose, smoking, hypertension). The educational attainment, socio-demographic and lifestyle behavior information were obtained through a structured self-report questionnaire, and Health-related quality of life (HRQoL) through the Short Form Survey (SF-12). Plasma Hcy and Lp(a) were determined in the fasting state. Results A CRS within the moderate-high risk range was reported in 21.7% donors. Multivariate logistic regression, after adjustment for clinical and demographic variables, showed that Hcy [OR (95% CI): 1.09 (1.04–1.13); p < 0.001) and low educational attainment [1.71 (1.09–2.73); p = 0.019] are independent risk factors for moderate-to-high CVD risk. Instead, Lp(a), evaluated in 774 donors, was > 30 mg/dL in 22.4% of the examined population, but without any significant correlation with CRS. Conclusions Our study highlights a previously unappreciated need for CV risk assessment in blood donors, which may include evaluation of educational attainment as a non-traditional risk marker.
Collapse
Affiliation(s)
- Francesco Vadini
- FIDAS (Italian Federation of Associations of Blood Donors), Pescara, Italy.,Psychoinfectivology Service, Infectious Disease Unit, Pescara General Hospital, Pescara, Italy
| | - Francesca Santilli
- Department of Medicine and Aging, "G. d'Annunzio" University of Chieti-Pescara, Chieti, Italy. .,Center of Aging Science and Translational Medicine (CESI-Met), "G. D'Annunzio" University Foundation, Via Luigi Polacchi, 66013, Chieti, Italy.
| | - Giuseppe Casalini
- Department of Hematology, Transfusion Medicine and Biotechnologies, Pescara General Hospital, Pescara, Italy
| | - Mario dell'Isola
- Department of Hematology, Transfusion Medicine and Biotechnologies, Pescara General Hospital, Pescara, Italy
| | - Ornella Iuliani
- Department of Hematology, Transfusion Medicine and Biotechnologies, Pescara General Hospital, Pescara, Italy
| | - Damiano D'Ardes
- Department of Medicine and Aging, "G. d'Annunzio" University of Chieti-Pescara, Chieti, Italy
| | - Luisa Lattanzio
- FIDAS (Italian Federation of Associations of Blood Donors), Pescara, Italy
| | - Marta Di Nicola
- Department of Medicine and Aging, "G. d'Annunzio" University of Chieti-Pescara, Chieti, Italy
| | | | - Patrizia Accorsi
- Department of Hematology, Transfusion Medicine and Biotechnologies, Pescara General Hospital, Pescara, Italy
| |
Collapse
|
17
|
Review of serum biomarkers in carotid atherosclerosis. J Vasc Surg 2019; 71:329-341. [PMID: 31327598 DOI: 10.1016/j.jvs.2019.04.488] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2018] [Accepted: 04/23/2019] [Indexed: 01/23/2023]
Abstract
BACKGROUND Carotid artery atherosclerotic stenosis is a preventable major cause of stroke, but there is still a need for definition of high-risk plaque in asymptomatic patients who might benefit from interventional therapies. Several image markers are recommended to characterize unstable plaques. The measurement of serum biomarkers is a promising method to assist in decision making, but the lack of robust evidence in the carotid environment burdens their potential as a standard of care. The goal of this review was to offer an updated state-of-the-art study of available serum biomarkers with clinical implications, with focus on those that may predict carotid symptom development. METHODS The Cochrane Library and MEDLINE databases were searched (all until September 2018) for studies on carotid plaque and serum biomarkers of atherosclerosis. Nonhuman, basic science, and histology studies were excluded, focusing on clinical studies. Selected abstracts were screened to include the most relevant articles on atherosclerotic plaque presence, progression, instability or symptom development. RESULTS Some well-established biomarkers for coronary disease are not relevant to carotid atherosclerosis and other inflammatory biomarkers, lipids, interleukins, homocysteine, and adipokines may be useful in quantifying carotid disease-related risk. Some serum biomarkers combined with image features may assist vascular specialists in selecting patients at high risk for stroke and in need of intervention. CONCLUSIONS Prospective studies applying a combination of biomarkers are essential to prove clinical usefulness.
Collapse
|
18
|
Wang Z, Zhai X, Xue M, Cheng W, Hu H. Prognostic value of lipoprotein (a) level in patients with coronary artery disease: a meta-analysis. Lipids Health Dis 2019; 18:150. [PMID: 31286992 PMCID: PMC6615167 DOI: 10.1186/s12944-019-1092-6] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2019] [Accepted: 06/26/2019] [Indexed: 12/31/2022] Open
Abstract
Background Elevated lipoprotein (a) is recognized as a risk factor for incident cardiovascular events in the general population and established cardiovascular disease patients. However, there are conflicting findings on the prognostic utility of elevated lipoprotein (a) level in patients with coronary artery disease (CAD).Thus, we performed a meta-analysis to evaluate the prognostic value of elevated lipoprotein (a) level in CAD patients. Methods and results A systematic literature search of PubMed and Embase databases was conducted until April 16, 2019. Observational studies reporting the prognostic value of elevated lipoprotein (a) level for cardiac events (cardiac death and acute coronary syndrome), cardiovascular events (death, stroke, acute coronary syndrome or coronary revascularisation), cardiovascular death, and all-cause mortality in CAD patients were included. Pooled multivariable adjusted risk ratio (RR) and 95% confidence interval (CI) for the highest vs. the lowest lipoprotein (a) level were utilized to calculate the prognostic value. Seventeen studies enrolling 283,328 patients were identified. Meta-analysis indicated that elevated lipoprotein (a) level was independently associated with an increased risk of cardiac events (RR 1.78; 95% CI 1.31–2.42) and cardiovascular events (RR 1.29; 95% CI 1.17–1.42) in CAD patients. However, elevated lipoprotein (a) level was not significantly associated with an increased risk of cardiovascular mortality (RR 1.43; 95% CI 0.94–2.18) and all-cause mortality (RR 1.35; 95% CI 0.93–1.95). Conclusions Elevated lipoprotein (a) level is an independent predictor of cardiac and cardiovascular events in CAD patients. Measurement of lipoprotein (a) level has potential to improve the risk stratification among patients with CAD. Electronic supplementary material The online version of this article (10.1186/s12944-019-1092-6) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Zhimiao Wang
- Department of Cardiology, Shandong Provincial Qianfoshan Hospital, the First Hospital Affiliated with Shandong First Medical University, NO. 16766, Jingshi Road, Jinan city, Jinan, 250014, Shandong Province, China
| | - Xincheng Zhai
- Department of Geriatrics, Municipal Hospital of Zibo City, Zibo City, 255000, Shandong Province, China
| | - Mei Xue
- Department of Cardiology, Shandong Provincial Qianfoshan Hospital, the First Hospital Affiliated with Shandong First Medical University, NO. 16766, Jingshi Road, Jinan city, Jinan, 250014, Shandong Province, China
| | - Wenjuan Cheng
- Department of Cardiology, Shandong Provincial Qianfoshan Hospital, the First Hospital Affiliated with Shandong First Medical University, NO. 16766, Jingshi Road, Jinan city, Jinan, 250014, Shandong Province, China
| | - Hesheng Hu
- Department of Cardiology, Shandong Provincial Qianfoshan Hospital, the First Hospital Affiliated with Shandong First Medical University, NO. 16766, Jingshi Road, Jinan city, Jinan, 250014, Shandong Province, China.
| |
Collapse
|
19
|
Fiuza M. Lipoprotein(a) as a novel therapeutic target for preventing cardiovascular disease: A whiter shade of pale? Rev Port Cardiol 2019; 38:495-496. [DOI: 10.1016/j.repc.2019.08.006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
|
20
|
Fiuza M. Lipoprotein(a) as a novel therapeutic target for preventing cardiovascular disease: A whiter shade of pale? REVISTA PORTUGUESA DE CARDIOLOGIA (ENGLISH EDITION) 2019. [DOI: 10.1016/j.repce.2019.09.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
|
21
|
Lin T, Wang L, Guo J, Liu P, Chen L, Wei M, Li G. Association Between Serum LDL-C and ApoB and SYNTAX Score in Patients With Stable Coronary Artery Disease. Angiology 2018; 69:724-729. [PMID: 29310455 DOI: 10.1177/0003319717748771] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The aim of this study was to examine the relationship between low-density lipoprotein cholesterol (LDL-C) and apolipoprotein (Apo) B levels and the SYNergy between percutaneous coronary intervention with TAXus and cardiac surgery (SYNTAX) score (SS) in patients with stable angina pectoris. We enrolled 594 patients who were suspected to have coronary heart disease (CHD) and who underwent coronary angiography. Patients were divided into 4 groups based on the SS: normal (SS = 0, n = 154), low SS (SS ≤ 22, n = 210), intermediate SS (22 < SS < 32, n = 122), and high SS (SS ≥ 33, n = 63). Positive correlations between lipoprotein (a), LDL-C, ApoB, total cholesterol, and SS were significant ( r = 0.132, 0.632, 0.599, and 0.313, respectively; P < .01), whereas high-density lipoprotein cholesterol (HDL-C), ApoA1, and ApoA1/ApoB levels showed a significant negative correlation ( r = -0.29, -0.344, and -0.561, respectively; P < .01). Multivariate linear regression analysis revealed that LDL-C, ApoB, ApoA1/ApoB, fibrinogen (Fg), and HDL-C levels had an effect on SS (standardized regression coefficients were 0.41, 0.29, -0.12, 0.08, and -0.09, respectively; P < .05). In conclusion, LDL-C, ApoB, ApoA1/ApoB, Fg, and HDL-C levels affected the SS and were predictors of CHD complexity.
Collapse
Affiliation(s)
- Taiwu Lin
- 1 Department of Cardiology, Heart Center, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Luzhao Wang
- 2 Department of Cardiology, Hanzhong Central Hospital, Hanzhong, China
| | - Jingbin Guo
- 1 Department of Cardiology, Heart Center, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Peng Liu
- 1 Department of Cardiology, Heart Center, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Liheng Chen
- 1 Department of Cardiology, Heart Center, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Mengqiu Wei
- 1 Department of Cardiology, Heart Center, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Gongxin Li
- 1 Department of Cardiology, Heart Center, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
22
|
Cicero AF, Bove M, Borghi C. Pharmacokinetics, pharmacodynamics and clinical efficacy of non-statin treatments for hypercholesterolemia. Expert Opin Drug Metab Toxicol 2017; 14:9-15. [DOI: 10.1080/17425255.2018.1416094] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Affiliation(s)
- Arrigo F.G. Cicero
- Medical and Surgical Sciences Department, University of Bologna, Bologna Italy
| | - Marilisa Bove
- Medical and Surgical Sciences Department, University of Bologna, Bologna Italy
| | - Claudio Borghi
- Medical and Surgical Sciences Department, University of Bologna, Bologna Italy
| |
Collapse
|
23
|
Corral P, Ruiz AJ. [PCSK-9 inhibitors, effects on LDL-C and future implications: What you should know]. HIPERTENSION Y RIESGO VASCULAR 2017; 34:176-183. [PMID: 28709786 DOI: 10.1016/j.hipert.2017.06.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2017] [Accepted: 06/01/2017] [Indexed: 10/19/2022]
Abstract
The discovery of proprotein convertase subtilisin/kexin type 9 (PCSK9) in 2003 in families with familial hypercholesterolemia (HF) later generated the development of pharmacological strategies in order to inhibit this protein. Twelve years after this discovery, the first two biological compounds (monoclonal antibodies) were approved, which have been shown to substantially decrease LDL-C and other lipid subfractions. The objective of the present article is to review the history of the discovery of PCSK9, its physiology and pathophysiology and subsequent pharmacological development. The objectives and goals reached to date and the pending questions regarding the efficacy and safety of its clinical use are presented.
Collapse
Affiliation(s)
- P Corral
- Especialista en Medicina Interna, lipidólogo clínico, Facultad Medicina, Universidad FASTA, Departamento Farmacología, Mar del Plata, Buenos Aires, Argentina.
| | - A J Ruiz
- Departamento de Medicina Interna, Departamento de Epidemiología Clínica y Bioestadística, Facultad de Medicina, Pontificia Universidad Javeriana, Bogotá D.C., Colombia
| |
Collapse
|
24
|
Pokrovsky SN, Afanasieva OI, Safarova MS, Balakhonova TV, Matchin YG, Adamova IYU, Konovalov GA, Ezhov MV. Specific Lp(a) apheresis: A tool to prove lipoprotein(a) atherogenicity. ATHEROSCLEROSIS SUPP 2017; 30:166-173. [PMID: 29096833 DOI: 10.1016/j.atherosclerosissup.2017.05.004] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND An elevated lipoprotein(a) (Lp(a)) level is observed in more than 30% of patients with stable ischemic heart disease (SIHD). We conducted an investigation of the effects of specific Lp(a) apheresis on the progression of atherosclerosis in SIHD patients with Lp(a) levels greater than 50 mg/dL. METHODS We prospectively enrolled 15 patients diagnosed with SIHD based on symptom-driven coronary angiography findings, with Lp(a) ≥50 mg/dL and a low density lipoprotein cholesterol (LDL-C) ≤2.5 mmol/L, who were on long-term statin therapy. They underwent weekly Lp(a) apheresis using Lp(a) Lipopak® adsorption columns which contain monospecific sheep polyclonal antibodies against human Lp(a). Fifteen age and gender matched SIHD patients receiving atorvastatin monotherapy served as controls. At baseline and 18 months post-treatment, quantitative coronary angiography, intracoronary ultrasound with virtual histology and carotid ultrasound were performed. Lipid profile, including Lp(a), was measured at the scheduled visits, and before and after each apheresis procedure. Levels of high-sensitivity C-reactive protein (hsCRP), matrix metalloproteinases (MMP)-7 and 9, and tissue inhibitor of matrix metalloproteinases (TIMP)-1 and 2 were determined at baseline and at the end of the study period. RESULTS Each specific Lp(a) apheresis procedure was carried out with two adsorption columns resulting in an average acute decrease in Lp(a) levels of 75% (from 110 ± 22 to 29 ± 16 mg/dL) without significant changes in other plasma components. Lp(a) reduction over the course of 18 months was associated with a decrease in the mean percent diameter stenosis of 5.05% and an increase in minimal lumen diameter of 14%; the mean total atheroma volume was reduced by 4.60 mm3 (p < 0.05 for all). There was a decrease in absolute common carotid intima-media thickness in the Lp(a) apheresis group of 0.07 ± 0.15 mm both from baseline and compared with the control group (p = 0.01). Levels of hsCRP were reduced by 40% in patients on Lp(a) apheresis without significant changes in the levels of other biomarkers at the end of the study. CONCLUSION Reduction of the atherosclerotic burden in coronary and carotid arteries was observed in patients treated with specific Lp(a) apheresis and statin over 18 months compared with statin therapy alone. These findings support the atherogenic role of Lp(a) and reinforce the need to assess the effects of Lp(a)-lowering on cardiovascular events and mortality. Trial Registration Clinicaltrials.gov (NCT02133807).
Collapse
Affiliation(s)
- S N Pokrovsky
- Laboratory of Atherosclerosis, Institute of Experimental Cardiology, Federal State Institution "Russian Cardiology Research and Production Center" of Ministry of Health of the Russian Federation, 15A, 3d Cherepkovskaya Street, Moscow 121552, Russia.
| | - O I Afanasieva
- Laboratory of Atherosclerosis, Institute of Experimental Cardiology, Federal State Institution "Russian Cardiology Research and Production Center" of Ministry of Health of the Russian Federation, 15A, 3d Cherepkovskaya Street, Moscow 121552, Russia
| | - M S Safarova
- Atherosclerosis Department, Institute of Clinical Cardiology named after A.L. Myasnikov, Federal State Institution "Russian Cardiology Research and Production Center" of Ministry of Health of the Russian Federation, 15A, 3d Cherepkovskaya Street, Moscow 121552, Russia
| | - T V Balakhonova
- Ultrasound Laboratory, Institute of Clinical Cardiology named after A.L. Myasnikov, Federal State Institution "Russian Cardiology Research and Production Center" of Ministry of Health of the Russian Federation, 15A, 3d Cherepkovskaya Street, Moscow 121552, Russia
| | - Yu G Matchin
- Catheterization Laboratory, Institute of Clinical Cardiology named after A.L. Myasnikov, Federal State Institution "Russian Cardiology Research and Production Center" of Ministry of Health of the Russian Federation, 15A, 3d Cherepkovskaya Street, Moscow 121552, Russia
| | - I Y U Adamova
- Laboratory of Atherosclerosis, Institute of Experimental Cardiology, Federal State Institution "Russian Cardiology Research and Production Center" of Ministry of Health of the Russian Federation, 15A, 3d Cherepkovskaya Street, Moscow 121552, Russia
| | - G A Konovalov
- Center of Extracorporeal Therapies, MEDSI Clinic, 3A, Georgian Lane, Moscow 123056, Russia
| | - M V Ezhov
- Atherosclerosis Department, Institute of Clinical Cardiology named after A.L. Myasnikov, Federal State Institution "Russian Cardiology Research and Production Center" of Ministry of Health of the Russian Federation, 15A, 3d Cherepkovskaya Street, Moscow 121552, Russia
| |
Collapse
|
25
|
Kersten E, Paun CC, Schellevis RL, Hoyng CB, Delcourt C, Lengyel I, Peto T, Ueffing M, Klaver CCW, Dammeier S, den Hollander AI, de Jong EK. Systemic and ocular fluid compounds as potential biomarkers in age-related macular degeneration. Surv Ophthalmol 2017; 63:9-39. [PMID: 28522341 DOI: 10.1016/j.survophthal.2017.05.003] [Citation(s) in RCA: 87] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2016] [Revised: 05/09/2017] [Accepted: 05/09/2017] [Indexed: 12/15/2022]
Abstract
Biomarkers can help unravel mechanisms of disease and identify new targets for therapy. They can also be useful in clinical practice for monitoring disease progression, evaluation of treatment efficacy, and risk assessment in multifactorial diseases, such as age-related macular degeneration (AMD). AMD is a highly prevalent progressive retinal disorder for which multiple genetic and environmental risk factors have been described, but the exact etiology is not yet fully understood. Many compounds have been evaluated for their association with AMD. We performed an extensive literature review of all compounds measured in serum, plasma, vitreous, aqueous humor, and urine of AMD patients. Over 3600 articles were screened, resulting in more than 100 different compounds analyzed in AMD studies, involved in neovascularization, immunity, lipid metabolism, extracellular matrix, oxidative stress, diet, hormones, and comorbidities (such as kidney disease). For each compound, we provide a short description of its function and discuss the results of the studies in relation to its usefulness as AMD biomarker. In addition, biomarkers identified by hypothesis-free techniques, including metabolomics, proteomics, and epigenomics, are covered. In summary, compounds belonging to the oxidative stress pathway, the complement system, and lipid metabolism are the most promising biomarker candidates for AMD. We hope that this comprehensive survey of the literature on systemic and ocular fluid compounds as potential biomarkers in AMD will provide a stepping stone for future research and possible implementation in clinical practice.
Collapse
Affiliation(s)
- Eveline Kersten
- Department of Ophthalmology, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Constantin C Paun
- Department of Ophthalmology, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Rosa L Schellevis
- Department of Ophthalmology, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Carel B Hoyng
- Department of Ophthalmology, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Cécile Delcourt
- Université de Bordeaux, ISPED, Bordeaux, France; INSERM, U1219-Bordeaux Population Health Research Center, Bordeaux, France
| | - Imre Lengyel
- Centre for Experimental Medicine, School of Medicine, Dentistry and Biomedical Science, Queen's University Belfast, Northern Ireland, United Kingdom
| | - Tunde Peto
- Centre for Public Health, School of Medicine, Dentistry and Biomedical Science, Queen's University Belfast, Northern Ireland, United Kingdom
| | - Marius Ueffing
- Department for Ophthalmology and Medical Bioanalytics Centre Tübingen, Institute for Ophthalmic Research, University of Tübingen, Tübingen, Germany
| | - Caroline C W Klaver
- Department of Ophthalmology, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen, the Netherlands; Department of Epidemiology, Erasmus Medical Center, Rotterdam, the Netherlands; Department of Ophthalmology, Erasmus Medical Center, Rotterdam, the Netherlands
| | - Sascha Dammeier
- Department for Ophthalmology and Medical Bioanalytics Centre Tübingen, Institute for Ophthalmic Research, University of Tübingen, Tübingen, Germany
| | - Anneke I den Hollander
- Department of Ophthalmology, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen, the Netherlands; Department of Human Genetics, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Eiko K de Jong
- Department of Ophthalmology, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen, the Netherlands.
| |
Collapse
|
26
|
Li S, Wu NQ, Zhu CG, Zhang Y, Guo YL, Gao Y, Li XL, Qing P, Cui CJ, Xu RX, Sun J, Liu G, Dong Q, Li JJ. Significance of lipoprotein(a) levels in familial hypercholesterolemia and coronary artery disease. Atherosclerosis 2017; 260:67-74. [PMID: 28351002 DOI: 10.1016/j.atherosclerosis.2017.03.021] [Citation(s) in RCA: 65] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2016] [Revised: 03/09/2017] [Accepted: 03/16/2017] [Indexed: 11/24/2022]
|
27
|
García-Gómez C, Martín-Martínez MA, Castañeda S, Sanchez-Alonso F, Uriarte-Ecenarro M, González-Juanatey C, Romera-Baures M, Santos-Rey J, Pinto-Tasende JA, Quesada-Masachs E, Tornero-Molina J, Martínez-González O, Cobo-Ibáñez T, Chamizo-Carmona E, Manrique-Arija S, Fábregas-Canales D, Díaz-González F, Llorca J, González-Gay MA. Lipoprotein(a) concentrations in rheumatoid arthritis on biologic therapy: Results from the CARdiovascular in rheuMAtology study project. J Clin Lipidol 2017; 11:749-756.e3. [PMID: 28476652 DOI: 10.1016/j.jacl.2017.02.018] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2016] [Revised: 12/22/2016] [Accepted: 02/24/2017] [Indexed: 10/20/2022]
Abstract
BACKGROUND Plasma concentrations of lipoprotein (a) (Lp(a)), a lipoprotein with atherogenic and thrombogenic properties, have a strong genetic basis, although high concentrations of Lp(a) have also been reported in the context of inflammation, as in rheumatoid arthritis (RA). Few studies evaluate the impact of biologic therapies (BT) on Lp(a) in RA, taking into account that with these new therapies a better control of inflammation is achieved. OBJECTIVE The aim of the study was to evaluate the plasma concentrations of Lp(a) in Spanish RA patients on BT attending rheumatology outpatient clinics. METHODS Baseline analysis of the CARdiovascular in rheuMAtology project, a 10-year prospective study, evaluating the risk of cardiovascular events in RA and other forms of inflammatory arthritis. RA patients were classified according to treatment: no biologic, anti-tumor necrosis factor, anti-interleukin-6 receptor tocilizumab (TCZ), and other biologic (rituximab or abatacept). A model of linear multivariate regression was built in which the dependent variable was Lp(a) concentration and the explanatory variable was BT. The model was adjusted for confounding factors. RESULTS Seven hundred and seventy-five RA patients were analyzed. Plasma concentrations of total cholesterol and triglyceride were significantly higher in TCZ-treated patients. Nevertheless, no significant difference in the atherogenic index between TCZ-treated patients and patients without BT was found. After adjusting for confounding factors, patients with BT had lower concentrations of Lp(a) than those without BT; however, only TCZ-treated patients achieved statistically significant differences (β: -0.303, 95% confidence interval: -0.558 to -0.047; P = .02). CONCLUSIONS RA patients treated with TCZ show lower plasma concentrations of Lp(a) compared with patients without BT.
Collapse
Affiliation(s)
- Carmen García-Gómez
- Division of Rheumatology, Consorci Sanitari de Terrassa, Terrassa, Barcelona, Spain
| | | | - Santos Castañeda
- Division of Rheumatology, Hospital U de la Princesa, IIS-IP, Madrid, Spain
| | | | | | | | - Montserrat Romera-Baures
- Division of Rheumatology, Hospital Universitari de Bellvitge, L'Hospitalet de Llobregat, Barcelona, Spain
| | - José Santos-Rey
- Division of Rheumatology, Hospital Virgen de la Salud, Toledo, Spain
| | | | | | - Jesús Tornero-Molina
- Division of Rheumatology, Hospital Universitario de Guadalajara, Guadalajara, Spain
| | | | - Tatiana Cobo-Ibáñez
- Division of Rheumatology, Hospital Universitario Infanta Sofía, San Sebastián de Los Reyes, Spain
| | | | - Sara Manrique-Arija
- Division of Rheumatology, Hospital Regional Universitario Carlos Haya, Málaga, Spain
| | | | - Federico Díaz-González
- Research Unit of Spanish Society of Rheumatology, Madrid, Spain; School of Medicine, Universidad de La Laguna, Tenerife, Spain; Division of Rheumatology, Hospital Universitario de Canarias, San Cristóbal de La Laguna, Santa Cruz de Tenerife, Spain
| | - Javier Llorca
- University of Cantabria - IDIVAL, and CIBER Epidemiologia y Salud Pública (CIBERESP), Santander, Spain
| | - Miguel A González-Gay
- Rheumatology Division, Epidemiology, Genetics and Atherosclerosis Research Group on Systemic Inflammatory Diseases, Hospital Universitario Marqués de Valdecilla, IDIVAL, University of Cantabria, Santander, Spain.
| | | |
Collapse
|
28
|
Schwartz J, Padmanabhan A, Aqui N, Balogun RA, Connelly-Smith L, Delaney M, Dunbar NM, Witt V, Wu Y, Shaz BH. Guidelines on the Use of Therapeutic Apheresis in Clinical Practice-Evidence-Based Approach from the Writing Committee of the American Society for Apheresis: The Seventh Special Issue. J Clin Apher 2017; 31:149-62. [PMID: 27322218 DOI: 10.1002/jca.21470] [Citation(s) in RCA: 276] [Impact Index Per Article: 39.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The American Society for Apheresis (ASFA) Journal of Clinical Apheresis (JCA) Special Issue Writing Committee is charged with reviewing, updating, and categorizing indications for the evidence-based use of therapeutic apheresis in human disease. Since the 2007 JCA Special Issue (Fourth Edition), the Committee has incorporated systematic review and evidence-based approaches in the grading and categorization of apheresis indications. This Seventh Edition of the JCA Special Issue continues to maintain this methodology and rigor to make recommendations on the use of apheresis in a wide variety of diseases/conditions. The JCA Seventh Edition, like its predecessor, has consistently applied the category and grading system definitions in the fact sheets. The general layout and concept of a fact sheet that was used since the fourth edition has largely been maintained in this edition. Each fact sheet succinctly summarizes the evidence for the use of therapeutic apheresis in a specific disease entity. The Seventh Edition discusses 87 fact sheets (14 new fact sheets since the Sixth Edition) for therapeutic apheresis diseases and medical conditions, with 179 indications, which are separately graded and categorized within the listed fact sheets. Several diseases that are Category IV which have been described in detail in previous editions and do not have significant new evidence since the last publication are summarized in a separate table. The Seventh Edition of the JCA Special Issue serves as a key resource that guides the utilization of therapeutic apheresis in the treatment of human disease. J. Clin. Apheresis 31:149-162, 2016. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Joseph Schwartz
- Department of Pathology and Cell Biology, Columbia University Medical Center, New York, New York
| | - Anand Padmanabhan
- Blood Center of Wisconsin, Department of Pathology, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Nicole Aqui
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Rasheed A Balogun
- Division of Nephrology, University of Virginia, Charlottesville, Virginia
| | - Laura Connelly-Smith
- Department of Medicine, Seattle Cancer Care Alliance and University of Washington, Seattle, Washington
| | - Meghan Delaney
- Bloodworks Northwest, Department of Laboratory Medicine, University of Washington, Seattle, Washington
| | - Nancy M Dunbar
- Department of Pathology and Laboratory Medicine, Dartmouth-Hitchcock Medical Center, Lebanon, New Hampshire
| | - Volker Witt
- Department for Pediatrics, St. Anna Kinderspital, Medical University of Vienna, Vienna, Austria
| | - Yanyun Wu
- Bloodworks Northwest, Department of Laboratory Medicine, Yale University School of Medicine, New Haven, Connecticut
| | - Beth H Shaz
- Department of Pathology and Cell Biology, Columbia University Medical Center, New York, New York.,New York Blood Center, Department of Pathology.,Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, Georgia
| |
Collapse
|
29
|
Alarcón García JC, Rodríguez Suárez S, Muñiz Grijalvo O, García Morillo S. Vascular lesions in patient with Takayasu arteritis and massive elevated lipoprotein(a) levels. Residual involvement or premature aterosclerosis? CLINICA E INVESTIGACION EN ARTERIOSCLEROSIS 2017; 29:98-102. [PMID: 28188021 DOI: 10.1016/j.arteri.2016.10.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/05/2016] [Revised: 10/22/2016] [Accepted: 10/28/2016] [Indexed: 02/05/2023]
Abstract
Lipoprotein (a) [Lp(a)] is a lipoprotein defined by presenting a specific apolipoprotein, ApoA, linked to the ApoB-100 by different types of chemical bonds, including a disulfide bridge. Despite their atherogenic mechanism is not fully understood, its importance has been demonstrated in the development of premature aterosclerosis. Multiple studies have shown its role as a cardiovascular risk factor associated with heart disease and stroke. We report the case of a patient with a diagnosis of Takayasu arteritis in which a massive elevation of Lp(a) was detected. We emphasize its diagnostic and therapeutic implications.
Collapse
Affiliation(s)
- José Carlos Alarcón García
- Unidad de Colagenosis y Enfermedades Minoritarias, Unidad Experimental de Riesgo Cardiovascular, Servicio de Medicina Interna, Hospital Universitario Virgen del Rocío, Sevilla, España.
| | - Santiago Rodríguez Suárez
- Unidad de Colagenosis y Enfermedades Minoritarias, Unidad Experimental de Riesgo Cardiovascular, Servicio de Medicina Interna, Hospital Universitario Virgen del Rocío, Sevilla, España
| | - Ovidio Muñiz Grijalvo
- Unidad de Colagenosis y Enfermedades Minoritarias, Unidad Experimental de Riesgo Cardiovascular, Servicio de Medicina Interna, Hospital Universitario Virgen del Rocío, Sevilla, España
| | - Salvador García Morillo
- Unidad de Colagenosis y Enfermedades Minoritarias, Unidad Experimental de Riesgo Cardiovascular, Servicio de Medicina Interna, Hospital Universitario Virgen del Rocío, Sevilla, España
| |
Collapse
|
30
|
Gaudet D, Watts GF, Robinson JG, Minini P, Sasiela WJ, Edelberg J, Louie MJ, Raal FJ. Effect of Alirocumab on Lipoprotein(a) Over ≥1.5 Years (from the Phase 3 ODYSSEY Program). Am J Cardiol 2017; 119:40-46. [PMID: 27793396 DOI: 10.1016/j.amjcard.2016.09.010] [Citation(s) in RCA: 86] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/28/2016] [Revised: 09/07/2016] [Accepted: 09/07/2016] [Indexed: 10/20/2022]
Abstract
Elevated lipoprotein(a) [Lp(a)] is independently associated with increased cardiovascular risk. However, treatment options for elevated Lp(a) are limited. Alirocumab, a monoclonal antibody to proprotein convertase subtilisin/kexin type 9, reduced low-density lipoprotein cholesterol (LDL-C) by up to 62% from baseline in phase 3 studies, with adverse event rates similar between alirocumab and controls. We evaluated the effect of alirocumab on serum Lp(a) using pooled data from the phase 3 ODYSSEY program: 4,915 patients with hypercholesterolemia from 10 phase 3 studies were included. Eight studies evaluated alirocumab 75 mg every 2 weeks (Q2W), with possible increase to 150 mg Q2W at week 12 depending on LDL-C at week 8 (75/150 mg Q2W); the other 2 studies evaluated alirocumab 150-mg Q2W from the outset. Comparators were placebo or ezetimibe. Eight studies were conducted on a background of statins, and 2 studies were carried out with no statins. Alirocumab was associated with significant reductions in Lp(a), regardless of starting dose and use of concomitant statins. At week 24, reductions from baseline were 23% to 27% with alirocumab 75/150-mg Q2W and 29% with alirocumab 150-mg Q2W (all comparisons p <0.0001 vs controls). Reductions were sustained over 78 to 104 weeks. Lp(a) reductions with alirocumab were independent of race, gender, presence of familial hypercholesterolemia, baseline Lp(a), and LDL-C concentrations, or use of statins. In conclusion, in addition to marked reduction in LDL-C, alirocumab leads to a significant and sustained lowering of Lp(a).
Collapse
|
31
|
Bertrand MJ, Tardif JC. Inflammation and beyond: new directions and emerging drugs for treating atherosclerosis. Expert Opin Emerg Drugs 2016; 22:1-26. [PMID: 27927063 DOI: 10.1080/14728214.2017.1269743] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
INTRODUCTION Cardiovascular (CV) atherosclerotic disease remains the leading cause of morbidity and mortality worldwide, despite the advances in contemporary therapies. Inflammation is an important process in atherosclerosis, leading to plaque rupture and acute coronary syndrome. Although statin therapy has substantially reduced CV events in primary and secondary prevention, many treated patients will have recurrent adverse CV events despite the standard of care. Thus, drug development aiming to target inflammatory pathways seems a promising avenue for novel therapies in atherosclerosis. Areas covered: Statins have been extensively studied and are the most effective lipid-lowering drugs available for CV prevention. Novel anti-inflammatory drugs are being tested in Phase II and III trials, targeting pathways like interleukin-1, leukotrienes, TNF-α, P-selectin, CCL2-CCR2 and MAP Kinase. Expert opinion: Novel anti-inflammatory therapies seem promising additions to address the residual CV risk present despite the current standard of care, but large clinical trials have not yet shown beneficial effects on clinical events. PCSK9 inhibitors have been shown to substantially reduce LDL-C, however their long-term safety and effects on CV risk are currently being investigated. Pharmacogenomics holds great potential in future lipid trials, enabling the identification of patients who will respond with greater benefits and smaller risk to therapies and to decrease failure rates in drug development, as genotype-dependent effects of the CETP inhibitor dalcetrapib were shown in the dal-OUTCOMES and dal-PLAQUE-2 trials.
Collapse
Affiliation(s)
- Marie-Jeanne Bertrand
- a Montreal Heart Institute, Department of medicine , Université de Montréal , Montreal , Canada
| | - Jean-Claude Tardif
- a Montreal Heart Institute, Department of medicine , Université de Montréal , Montreal , Canada
| |
Collapse
|
32
|
Bucci M, Tana C, Giamberardino MA, Cipollone F. Lp(a) and cardiovascular risk: Investigating the hidden side of the moon. Nutr Metab Cardiovasc Dis 2016; 26:980-986. [PMID: 27514608 DOI: 10.1016/j.numecd.2016.07.004] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/29/2016] [Revised: 06/21/2016] [Accepted: 07/06/2016] [Indexed: 12/21/2022]
Abstract
AIMS This article reports current evidence on the association between Lp(a) and cardiovascular (CV) disease and on pathophysiological mechanisms. The available information on therapy for reduction of lipoprotein(a) is also discussed. DATA SYNTHESIS Although some evidence is conflicting, Lp(a) seems to increase CV risk through stimulation of platelet aggregation, inhibition of tissue factor pathway inhibitor, alteration of fibrin clot structure and promotion of endothelial dysfunction and phospholipid oxidation. Lp(a) 3.5-fold higher than normal increases the risk of coronary heart disease and general CV events, particularly in those with LDL cholesterol ≥ 130 mg/dl. High Lp(a) values represent also an independent risk factor for ischemic stroke (more relevant in young stroke patients), peripheral artery disease (PAD) and aortic and mitral stenosis. Furthermore, high Lp(a) levels seem to be associated with increased risk of cardiovascular events in patients with chronic kidney disease, particularly in those undergoing percutaneous coronary intervention. CONCLUSIONS Lipoprotein (a) (Lp[a]) seems to significantly influence the risk of cardiovascular events. The effects of statins and fibrates on Lp(a) are limited and extremely variable. Nicotinic acid was shown effective in reducing Lp(a) but, due to its side effects and serious adverse events during clinical trials, it is no longer considered a possible option for treatment. To date, the treatment of choice for high levels of Lp(a) in high CV risk patients is represented by LDL-Apheresis. Thanks to innovative technologies, new selectively inhibiting LPA drugs are being developed and tested.
Collapse
Affiliation(s)
- M Bucci
- Regional Center for the Study of Atherosclerosis, Hypertension and Dyslipidemia, "SS Annunziata" Hospital - ASL Chieti, Italy; Ce.S.I.-Met, "G. D'Annunzio" University of Chieti, Italy
| | - C Tana
- Internal Medicine Unit, Guastalla Hospital, AUSL Reggio Emilia, Italy
| | - M A Giamberardino
- Ce.S.I.-Met, "G. D'Annunzio" University of Chieti, Italy; Geriatrics Clinic, Department of Medicine and Science of Aging, "G. D'Annunzio" University of Chieti, Italy
| | - F Cipollone
- Regional Center for the Study of Atherosclerosis, Hypertension and Dyslipidemia, "SS Annunziata" Hospital - ASL Chieti, Italy; Ce.S.I.-Met, "G. D'Annunzio" University of Chieti, Italy; Geriatrics Clinic, Department of Medicine and Science of Aging, "G. D'Annunzio" University of Chieti, Italy.
| |
Collapse
|
33
|
Yang SH, Li S, Zhang Y, Xu RX, Zhu CG, Guo YL, Wu NQ, Qing P, Gao Y, Cui CJ, Dong Q, Sun J, Li JJ. Analysis of the association between plasma PCSK9 and Lp(a) in Han Chinese. J Endocrinol Invest 2016; 39:875-83. [PMID: 26894681 DOI: 10.1007/s40618-016-0433-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2015] [Accepted: 01/20/2016] [Indexed: 01/04/2023]
Abstract
PURPOSE It has been reported that proprotein convertase subtilisin/kexin type 9 (PCSK9) inhibitors can significantly reduce lipoprotein(a) [Lp(a)], and the mechanism for Lp(a) reduction remains unclear. Recently an interesting clinical research with a small sample showed a positive correlation between plasma PCSK9 and Lp(a) levels in diabetes. Here we aimed to use a relatively large sample to investigate whether such an association exists in Han Chinese. METHODS A total of 783 inpatients were consecutively enrolled and composed of 172 patients with type 2 diabetes mellitus (T2DM) and 611 non-T2DM subjects. Plasma PCSK9 level was measured by ELISA, and its association with Lp(a) was assayed by Spearman's correlation and multiple regression. Clinical and biochemical parameters were determined in all subjects studied. RESULTS No significant differences in PCSK9 and Lp(a) levels were found between T2DM and non-T2DM patients. PCSK9 level was not related to Lp(a) level either in T2DM or non-T2DM group in bivariate correlation and multiple linear regression analysis. Additionally, no association between the levels of PCSK9 and Lp(a) was found in well, poorly controlled T2DM patients or in T2DM patients with or without coronary artery disease (CAD). Besides, no difference was found among the PCSK9 values across tertiles of Lp(a) level. CONCLUSION We found no association of plasma PCSK9 levels with Lp(a) level in Han Chinese with or without T2DM, suggesting that Lp(a) reduction by PCSK9 inhibitors may not be achieved simply through PCSK9 pathway at least in Chinese.
Collapse
Affiliation(s)
- S-H Yang
- Division of Dyslipidemia, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Peking Union Medical College, Chinese Academy of Medical Sciences, BeiLiShi Road 167, Beijing, 100037, China
| | - S Li
- Division of Dyslipidemia, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Peking Union Medical College, Chinese Academy of Medical Sciences, BeiLiShi Road 167, Beijing, 100037, China
| | - Y Zhang
- Division of Dyslipidemia, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Peking Union Medical College, Chinese Academy of Medical Sciences, BeiLiShi Road 167, Beijing, 100037, China
| | - R-X Xu
- Division of Dyslipidemia, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Peking Union Medical College, Chinese Academy of Medical Sciences, BeiLiShi Road 167, Beijing, 100037, China
| | - C-G Zhu
- Division of Dyslipidemia, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Peking Union Medical College, Chinese Academy of Medical Sciences, BeiLiShi Road 167, Beijing, 100037, China
| | - Y-L Guo
- Division of Dyslipidemia, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Peking Union Medical College, Chinese Academy of Medical Sciences, BeiLiShi Road 167, Beijing, 100037, China
| | - N-Q Wu
- Division of Dyslipidemia, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Peking Union Medical College, Chinese Academy of Medical Sciences, BeiLiShi Road 167, Beijing, 100037, China
| | - P Qing
- Division of Dyslipidemia, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Peking Union Medical College, Chinese Academy of Medical Sciences, BeiLiShi Road 167, Beijing, 100037, China
| | - Y Gao
- Division of Dyslipidemia, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Peking Union Medical College, Chinese Academy of Medical Sciences, BeiLiShi Road 167, Beijing, 100037, China
| | - C-J Cui
- Division of Dyslipidemia, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Peking Union Medical College, Chinese Academy of Medical Sciences, BeiLiShi Road 167, Beijing, 100037, China
| | - Q Dong
- Division of Dyslipidemia, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Peking Union Medical College, Chinese Academy of Medical Sciences, BeiLiShi Road 167, Beijing, 100037, China
| | - J Sun
- Division of Dyslipidemia, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Peking Union Medical College, Chinese Academy of Medical Sciences, BeiLiShi Road 167, Beijing, 100037, China
| | - J-J Li
- Division of Dyslipidemia, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Peking Union Medical College, Chinese Academy of Medical Sciences, BeiLiShi Road 167, Beijing, 100037, China.
| |
Collapse
|
34
|
Katsiki N, Mikhailidis DP, Mantzoros CS. Non-alcoholic fatty liver disease and dyslipidemia: An update. Metabolism 2016; 65:1109-23. [PMID: 27237577 DOI: 10.1016/j.metabol.2016.05.003] [Citation(s) in RCA: 398] [Impact Index Per Article: 49.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2016] [Revised: 05/02/2016] [Accepted: 05/05/2016] [Indexed: 11/21/2022]
Abstract
Non-alcoholic fatty liver (NAFLD) is the most common liver disease worldwide, progressing from simple steatosis to necroinflammation and fibrosis (leading to non-alcoholic steatohepatitis, NASH), and in some cases to cirrhosis and hepatocellular carcinoma. Inflammation, oxidative stress and insulin resistance are involved in NAFLD development and progression. NAFLD has been associated with several cardiovascular (CV) risk factors including obesity, dyslipidemia, hyperglycemia, hypertension and smoking. NAFLD is also characterized by atherogenic dyslipidemia, postprandial lipemia and high-density lipoprotein (HDL) dysfunction. Most importantly, NAFLD patients have an increased risk for both liver and CV disease (CVD) morbidity and mortality. In this narrative review, the associations between NAFLD, dyslipidemia and vascular disease in NAFLD patients are discussed. NAFLD treatment is also reviewed with a focus on lipid-lowering drugs. Finally, future perspectives in terms of both NAFLD diagnostic biomarkers and therapeutic targets are considered.
Collapse
Affiliation(s)
- Niki Katsiki
- Second Propedeutic Department of Internal Medicine, Medical School, Aristotle University of Thessaloniki, Hippocration Hospital, Thessaloniki, Greece
| | - Dimitri P Mikhailidis
- Department of Clinical Biochemistry (Vascular Disease Prevention Clinics), Royal Free Hospital Campus, University College London Medical School, University College London (UCL), London, UK.
| | - Christos S Mantzoros
- Division of Endocrinology, Diabetes and Metabolism, Department of Internal Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
35
|
Cho JH, Lee DY, Lee ES, Kim J, Park SE, Park CY, Lee WY, Oh KW, Park SW, Rhee EJ. Increased risk of coronary artery calcification progression in subjects with high baseline Lp(a) levels: The Kangbuk Samsung Health Study. Int J Cardiol 2016; 222:233-237. [PMID: 27497100 DOI: 10.1016/j.ijcard.2016.07.219] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/24/2016] [Accepted: 07/28/2016] [Indexed: 11/24/2022]
Abstract
BACKGROUND Results from previous studies support the association of lipoprotein(a) [Lp(a)] levels and coronary artery disease risk. In this study, we analyzed the association between baseline Lp(a) levels and future progression of coronary artery calcification (CAC) in apparently healthy Korean adults. METHODS A total of 2611 participants (mean age: 41years, 92% mend) who underwent a routine health check-up in 2010 and 2014 were enrolled. Coronary artery calcium score (CACS) were measured by multi-detector computed tomography. Baseline Lp(a) was measured by high-sensitivity immunoturbidimetric assay. Progression of CAC was defined as a change in CACS >0 over four years. RESULTS Bivariate correlation analyses with baseline Lp(a) and other metabolic parameters revealed age, total cholesterol, HDL-C, LDL-C and CACS to have a significant positive correlation, while body weight, fasting glucose level, blood pressure and triglyceride level were negatively correlated with baseline Lp(a) level. After four years of follow-up, 635 subjects (24.3%) had CAC progression. The participants who had CAC progression were older, composed of more men, more obese, and had higher fasting glucose levels and worse baseline lipid profiles compared to those who did not have CAC progression. The mean serum Lp(a) level was significantly higher in subjects who had CAC progression compared to those who did not (32.5 vs. 28.9mg/dL, p<0.01). When the risk for CAC progression according to baseline Lp(a) was calculated, those with Lp(a) level≥50mg/dL had an odds ratio of 1.333 (95% CI 1.027-1.730) for CAC progression compared to those with Lp(a)<50mg/dL after adjusting for confounding factors. CONCLUSIONS In this study, the subjects who had higher Lp(a) were at significantly higher risk for CAC progression after four years of follow-up, suggesting the role of high Lp(a) in CAC progression.
Collapse
Affiliation(s)
- Jung Hwan Cho
- Department of Endocrinology and Metabolism, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Da Young Lee
- Department of Endocrinology and Metabolism, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Eun Seo Lee
- Department of Endocrinology and Metabolism, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Jihyun Kim
- Department of Endocrinology and Metabolism, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Se Eun Park
- Department of Endocrinology and Metabolism, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Cheol-Young Park
- Department of Endocrinology and Metabolism, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Won-Young Lee
- Department of Endocrinology and Metabolism, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Ki-Won Oh
- Department of Endocrinology and Metabolism, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Sung-Woo Park
- Department of Endocrinology and Metabolism, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Eun-Jung Rhee
- Department of Endocrinology and Metabolism, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea.
| |
Collapse
|
36
|
Herrington W, Lacey B, Sherliker P, Armitage J, Lewington S. Epidemiology of Atherosclerosis and the Potential to Reduce the Global Burden of Atherothrombotic Disease. Circ Res 2016; 118:535-46. [PMID: 26892956 DOI: 10.1161/circresaha.115.307611] [Citation(s) in RCA: 879] [Impact Index Per Article: 109.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Atherosclerosis is a leading cause of vascular disease worldwide. Its major clinical manifestations include ischemic heart disease, ischemic stroke, and peripheral arterial disease. In high-income countries, there have been dramatic declines in the incidence and mortality from ischemic heart disease and ischemic stroke since the middle of the 20th century. For example, in the United Kingdom, the probability of death from vascular disease in middle-aged men (35-69 years) has decreased from 22% in 1950 to 6% in 2010. Most low- and middle-income countries have also reported declines in mortality from stroke over the last few decades, but mortality trends from ischemic heart disease have been more varied, with some countries reporting declines and others reporting increases (particularly those in Eastern Europe and Asia). Many major modifiable risk factors for atherosclerosis have been identified, and the causal relevance of several risk factors is now well established (including, but not limited to, smoking, adiposity, blood pressure, blood cholesterol, and diabetes mellitus). Widespread changes in health behaviors and use of treatments for these risk factors are responsible for some of the dramatic declines in vascular mortality in high-income countries. In order that these declines continue and are mirrored in less wealthy nations, increased efforts are needed to tackle these major risk factors, particularly smoking and the emerging obesity epidemic.
Collapse
Affiliation(s)
- William Herrington
- From the Clinical Trial Service Unit and Epidemiological Studies Unit (CTSU), Nuffield Department of Population Health, Oxford OX3 7LF, UK
| | - Ben Lacey
- From the Clinical Trial Service Unit and Epidemiological Studies Unit (CTSU), Nuffield Department of Population Health, Oxford OX3 7LF, UK
| | - Paul Sherliker
- From the Clinical Trial Service Unit and Epidemiological Studies Unit (CTSU), Nuffield Department of Population Health, Oxford OX3 7LF, UK
| | - Jane Armitage
- From the Clinical Trial Service Unit and Epidemiological Studies Unit (CTSU), Nuffield Department of Population Health, Oxford OX3 7LF, UK.
| | - Sarah Lewington
- From the Clinical Trial Service Unit and Epidemiological Studies Unit (CTSU), Nuffield Department of Population Health, Oxford OX3 7LF, UK
| |
Collapse
|
37
|
HyperLp(a)lipoproteinaemia: unmet need of diagnosis and treatment? BLOOD TRANSFUSION = TRASFUSIONE DEL SANGUE 2016; 14:408-12. [PMID: 27416577 DOI: 10.2450/2016.0027-16] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
38
|
Kotani K, Serban MC, Penson P, Lippi G, Banach M. Evidence-based assessment of lipoprotein(a) as a risk biomarker for cardiovascular diseases - Some answers and still many questions. Crit Rev Clin Lab Sci 2016; 53:370-8. [PMID: 27173621 DOI: 10.1080/10408363.2016.1188055] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
The present article is aimed at outlining the current state of knowledge regarding the clinical value of lipoprotein(a) (Lp(a)) as a marker of cardiovascular disease (CVD) risk by summarizing the results of recent clinical studies, meta-analyses and systematic reviews. The literature supports the predictive value of Lp(a) on CVD outcomes, although the effect size is modest. Lp(a) would also appear to have an effect on cerebrovascular outcomes, however the effect appears even smaller than that for CVD outcomes. Consideration of apolipoprotein(a) (apo(a)) isoforms and LPA genetics in relation to the simple assessment of Lp(a) concentration may enhance clinical practice in vascular medicine. We also describe recent advances in Lp(a) research (including therapies) and highlight areas where further research is needed such as the measurement of Lp(a) and its involvement in additional pathophysiological processes.
Collapse
Affiliation(s)
- Kazuhiko Kotani
- a Division of Community and Family MedicinevJichi Medical University , Shimotsuke-City , Japan .,b Department of Clinical Laboratory Medicine , Jichi Medical University , Shimotsuke-City , Japan
| | - Maria-Corina Serban
- c Department of Epidemiology , University of Alabama at Birmingham , Birmingham , AL , USA .,d Department of Functional Sciences , Discipline of Pathophysiology, "Victor Babes" University of Medicine and Pharmacy , Timisoara , Romania
| | - Peter Penson
- e Section of Clinical Biochemistry , School of Pharmacy and Biomolecular Sciences, Liverpool John Moores University , Liverpool , UK
| | - Giuseppe Lippi
- f Section of Clinical Biochemistry , University of Verona , Verona , Italy , and
| | - Maciej Banach
- g Department of Hypertension , Chair of Nephrology and Hypertension, Medical University of Lodz , Lodz , Poland
| |
Collapse
|
39
|
Acute Disseminated Encephalomyelitis. J Clin Apher 2016; 31:163-202. [PMID: 27322219 DOI: 10.1002/jca.21474] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
|
40
|
Matney K, Berg M, Falko JM, Draper NL. The role of lipoprotein(a) in clotting reactions during lipoprotein apheresis--A case report. J Clin Lipidol 2016; 10:438-42. [PMID: 27055976 DOI: 10.1016/j.jacl.2015.12.028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2015] [Revised: 12/22/2015] [Accepted: 12/26/2015] [Indexed: 11/26/2022]
Abstract
In individuals with familial hypercholesterolemia (FH) who are unable to reach a target low-density lipoprotein level on a drug regimen, lipoprotein apheresis (LA) may be the treatment of choice. Severe reactions involving clotting during LA are not well described in the literature. We report a case of a 63-year-old woman with FH and markedly elevated lipoprotein(a) (Lp[a]) levels who experienced such a reaction while undergoing LA with a dextran-sulfate cellulose column on the Kaneka MA-01 Liposorber system. Owing to the clotting as well as a blood pressure drop to <100 mm Hg systolic, the procedure was stopped early. Before her second procedure, she was given an increased loading dose of unfractionated heparin. She did not develop clotting during this second procedure. A growing body of literature on the role of Lp(a) in atherothrombotic complications and hemostasis supports a possible mechanism by which clotting in the instrument could occur during apheresis. Our patient's initial pretreatment Lp(a) was 3.5 times greater than the mean Lp(a) levels in patients with FH. This theory is consistent with our case in that the patient's Lp(a) levels progressively declined with each procedure, and she had no subsequent clotting.
Collapse
Affiliation(s)
- Kathryn Matney
- Department of Pathology, University of Colorado, Aurora, CO, USA.
| | - Mary Berg
- Department of Pathology, University of Colorado, Aurora, CO, USA
| | - James M Falko
- Department of Internal Medicine, University of Colorado, Aurora, CO, USA
| | - Nicole L Draper
- Department of Pathology, University of Colorado, Aurora, CO, USA
| |
Collapse
|
41
|
Streja D. Editorial: collecting new external evidence for cholesterol management. Curr Opin Endocrinol Diabetes Obes 2016; 23:121-3. [PMID: 26863279 DOI: 10.1097/med.0000000000000244] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Affiliation(s)
- Dan Streja
- David Geffen School of Medicine, University of California, Los Angeles, California, USA
| |
Collapse
|
42
|
Abstract
PURPOSE OF REVIEW Novel therapies for severe dyslipidemia target a wide range of unmet medical needs: severe familial hypercholesterolemia, severe hypertriglyceridemia and chylomicronemia, elevated lipoprotein (a), lipodystrophies, high-density lipoprotein particle diseases, lysosomal acid lipase deficiency and storage diseases, nonalcoholic fatty liver disease and others. The purpose of this review is to describe the contribution of human genetics to the development of therapeutic approaches targeting severe dyslipidemia. RECENT FINDINGS Recent advances in human genetics and the identification of rare genetic variants having strong effects on disease risk not only accelerated the development of therapies for severe dyslipidemia, they also revealed new pathways, genes and mechanisms of health, disease or drug response, and facilitated molecular diagnosis, which may prove essential as the authorized use of some of these novel drugs is limited to specific conditions. In addition, the dissection of the gene and cell machinery gave rise to new technologies, gene-based therapies and biodrugs covering a broad range of novel agents currently available or in clinical development to treat severe lipid disorders. SUMMARY Several novel therapies are recently available or under development to treat severe dyslipidemia and associated risk stem directly from genetic research. Altogether, these therapies target a broad variety of severe dyslipidemia pathways or mechanisms and illustrate that clinical lipidology has now entered the era of precision medicine.
Collapse
Affiliation(s)
- Daniel Gaudet
- Department of Medicine, Lipidology Unit, Community Genomic Medicine Center, Université de Montréal and ECOGENE-21 Clinical and Translational Research Center, Chicoutimi, Quebec, Canada
| |
Collapse
|
43
|
Saeedi R, Frohlich J. Lipoprotein (a), an independent cardiovascular risk marker. Clin Diabetes Endocrinol 2016; 2:7. [PMID: 28702242 PMCID: PMC5471681 DOI: 10.1186/s40842-016-0024-x] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2015] [Accepted: 03/10/2016] [Indexed: 01/05/2023] Open
Abstract
Epidemiological and genetic studies have identified elevated levels of lipoprotein (a) ((Lp(a)) as a causal and independent risk factor for cardiovascular diseases (CVD). The Lp(a)-induced increased risk of CVD may be mediated by both its proatherogenic and prothrombotic mechanisms. Several guidelines recommend screening of Lp(a) level; however, there are few treatment options for the management of patients with elevated Lp(a). Several new medications for Lp(a) are under development. PCSK9 inhibitors, apolipoprotein (a)-antisense, and apolipoprotein(B-100)-antisense mipomersen have shown promising results. Lp(a) reduction will continue to be an active area of investigation.
Collapse
Affiliation(s)
- Ramesh Saeedi
- Healthy Heart Program, St. Paul's Hospital, Rm 180-1081 Burrard Street, Vancouver, B.C V6Z 1Y6 Canada.,Pathology and Laboratory Medicine Department, University of British Columbia, St. Paul's Hospital, Rm 180-1081 Burrard Street, Vancouver, B.C V6Z 1Y6 Canada
| | - Jiri Frohlich
- Healthy Heart Program, St. Paul's Hospital, Rm 180-1081 Burrard Street, Vancouver, B.C V6Z 1Y6 Canada.,Pathology and Laboratory Medicine Department, University of British Columbia, St. Paul's Hospital, Rm 180-1081 Burrard Street, Vancouver, B.C V6Z 1Y6 Canada
| |
Collapse
|
44
|
Shantha GPS, Robinson JG. Emerging innovative therapeutic approaches targeting PCSK9 to lower lipids. Clin Pharmacol Ther 2015; 99:59-71. [DOI: 10.1002/cpt.281] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2015] [Accepted: 10/16/2015] [Indexed: 12/16/2022]
Affiliation(s)
- GPS Shantha
- Departments of Epidemiology & Medicine, Prevention Intervention Center, Department of Epidemiology, College of Public Health; University of Iowa; Iowa City Iowa USA
| | - JG Robinson
- Departments of Epidemiology & Medicine, Prevention Intervention Center, Department of Epidemiology, College of Public Health; University of Iowa; Iowa City Iowa USA
| |
Collapse
|