1
|
Zhu LR, He XH, Yuan YH, Yuan H, Xia XH. [Changes and significance of oxidized phospholipids and endothelial nitric oxide synthase in the acute stage of Kawasaki disease]. ZHONGGUO DANG DAI ER KE ZA ZHI = CHINESE JOURNAL OF CONTEMPORARY PEDIATRICS 2024; 26:829-834. [PMID: 39148387 PMCID: PMC11334547 DOI: 10.7499/j.issn.1008-8830.2403056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Accepted: 07/04/2024] [Indexed: 08/17/2024]
Abstract
OBJECTIVES To investigate the changes in the serum levels of oxidized phospholipids (OxPLs) and endothelial nitric oxide synthase (eNOS) and their association with coronary artery disease (CAL) in children in the acute stage of Kawasaki disease (KD), as well as the clinical significance of OxPLs and eNOS. METHODS A prospective study was conducted on 95 children in the acute stage of KD (KD group). According to the presence of absence of CAL, the KD group was further divided into a CAL subgroup and a non-CAL (NCAL) subgroup. Thirty children with fever due to lower respiratory tract infection were enrolled as the fever group. Thirty healthy children who underwent physical examination were enrolled as the healthy control group. The above groups were compared in terms of general information and serum levels of OxPLs, eNOS and other laboratory indexes, and the correlation between OxPLs level and eNOS level was analyzed. RESULTS The KD group had a significantly higher level of OxPLs and a significantly lower level of eNOS compared with the fever group and the healthy control group (P<0.05). After treatment, the children with KD had a significantly decreased OxPLs level and a significantly increased eNOS level (P<0.05). Compared with the NCAL subgroup, the CAL subgroup had a significantly higher level of OxPLs and a significantly lower level of eNOS (P<0.05). Among the children of KD, the level of OxPLs was negatively correlated with that of eNOS (rs=-0.353, P<0.05). CONCLUSIONS Serum OxPLs and eNOS in the acute stage of KD may be involved in the development of CAL in children with KD, and therefore, they may be used as the biomarkers to predict CAL in these children.
Collapse
Affiliation(s)
- Liu-Rong Zhu
- Department of Pediatrics, First Affiliated Hospital of Hunan Normal University, Changsha 410005, China
| | - Xue-Hua He
- Department of Pediatrics, First Affiliated Hospital of Hunan Normal University, Changsha 410005, China
| | - Yong-Hua Yuan
- Department of Pediatrics, First Affiliated Hospital of Hunan Normal University, Changsha 410005, China
| | | | | |
Collapse
|
2
|
Groenen AG, Matveyenko A, Matienzo N, Halmos B, Zhang H, Westerterp M, Reyes-Soffer G. Apolipoprotein(a) production and clearance are associated with plasma IL-6 and IL-18 levels, dependent on ethnicity. Atherosclerosis 2024; 391:117474. [PMID: 38428286 DOI: 10.1016/j.atherosclerosis.2024.117474] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Revised: 01/31/2024] [Accepted: 02/01/2024] [Indexed: 03/03/2024]
Abstract
BACKGROUND AND AIMS High plasma lipoprotein (a) [Lp(a)] levels are associated with increased atherosclerotic cardiovascular disease (ASCVD), in part attributed to elevated inflammation. High plasma Lp(a) levels inversely correlate with apolipoprotein (a) [(APO(a)] isoform size. APO(a) isoform size is negatively associated with APO(a) production rate (PR) and positively associated with APO(a) fractional catabolic rate (FCR). We asked whether APO(a) PR and FCR (kinetics) are associated with plasma levels of interleukin (IL)-6 and IL-18, pro-inflammatory interleukins that promote ASCVD. METHODS We used samples from existing data of APO(a) kinetic studies from an ethnically diverse cohort (n = 25: 10 Black, 9 Hispanic, and 6 White subjects) and assessed IL-6 and IL-18 plasma levels. We performed multivariate linear regression analyses to examine the relationships between predictors APO(a) PR or APO(a) FCR, and outcome variables IL-6 or IL-18. In these analyses, we adjusted for parameters known to affect Lp(a) levels and APO(a) PR and FCR, including race/ethnicity and APO(a) isoform size. RESULTS APO(a) PR and FCR were positively associated with plasma IL-6, independent of isoform size, and dependent on race/ethnicity. APO(a) PR was positively associated with plasma IL-18, independent of isoform size and race/ethnicity. APO(a) FCR was not associated with plasma IL-18. CONCLUSIONS Our studies demonstrate a relationship between APO(a) PR and FCR and plasma IL-6 or IL-18, interleukins that promote ASCVD. These studies provide new insights into Lp(a) pro-inflammatory properties and are especially relevant in view of therapies targeting APO(a) to decrease cardiovascular risk.
Collapse
Affiliation(s)
- Anouk G Groenen
- Department of Pediatrics, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Anastasiya Matveyenko
- Columbia University Irving Medical Center, College of Physicians and Surgeons, Department of Medicine, Division of Preventive Medicine and Nutrition, New York, NY, USA
| | - Nelsa Matienzo
- Columbia University Irving Medical Center, College of Physicians and Surgeons, Department of Medicine, Division of Preventive Medicine and Nutrition, New York, NY, USA
| | - Benedek Halmos
- Department of Pediatrics, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Hanrui Zhang
- Columbia University Irving Medical Center, Division of Cardiology, New York, NY, USA
| | - Marit Westerterp
- Department of Pediatrics, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands.
| | - Gissette Reyes-Soffer
- Columbia University Irving Medical Center, College of Physicians and Surgeons, Department of Medicine, Division of Preventive Medicine and Nutrition, New York, NY, USA.
| |
Collapse
|
3
|
Abstract
Prolonged or excessive exposure to oxidized phospholipids (OxPLs) generates chronic inflammation. OxPLs are present in atherosclerotic lesions and can be detected in plasma on apolipoprotein B (apoB)-containing lipoproteins. When initially conceptualized, OxPL-apoB measurement in plasma was expected to reflect the concentration of minimally oxidized LDL, but, surprisingly, it correlated more strongly with plasma lipoprotein(a) (Lp(a)) levels. Indeed, experimental and clinical studies show that Lp(a) particles carry the largest fraction of OxPLs among apoB-containing lipoproteins. Plasma OxPL-apoB levels provide diagnostic information on the presence and extent of atherosclerosis and improve the prognostication of peripheral artery disease and first and recurrent myocardial infarction and stroke. The addition of OxPL-apoB measurements to traditional cardiovascular risk factors improves risk reclassification, particularly in patients in intermediate risk categories, for whom improving decision-making is most impactful. Moreover, plasma OxPL-apoB levels predict cardiovascular events with similar or greater accuracy than plasma Lp(a) levels, probably because this measurement reflects both the genetics of elevated Lp(a) levels and the generalized or localized oxidation that modifies apoB-containing lipoproteins and leads to inflammation. Plasma OxPL-apoB levels are reduced by Lp(a)-lowering therapy with antisense oligonucleotides and by lipoprotein apheresis, niacin therapy and bariatric surgery. In this Review, we discuss the role of role OxPLs in the pathophysiology of atherosclerosis and Lp(a) atherogenicity, and the use of OxPL-apoB measurement for improving prognosis, risk reclassification and therapeutic interventions.
Collapse
Affiliation(s)
- Sotirios Tsimikas
- Division of Cardiovascular Medicine, University of California San Diego, La Jolla, CA, USA.
| | - Joseph L Witztum
- Division of Endocrinology and Metabolism, University of California San Diego, La Jolla, CA, USA
| |
Collapse
|
4
|
Chen S, Gao JJ, Liu YJ, Mo ZW, Wu FY, Hu ZJ, Peng YM, Zhang XQ, Ma ZS, Liu ZL, Yan JY, Ou ZJ, Li Y, Ou JS. The oxidized phospholipid PGPC impairs endothelial function by promoting endothelial cell ferroptosis via FABP3. J Lipid Res 2024; 65:100499. [PMID: 38218337 PMCID: PMC10864338 DOI: 10.1016/j.jlr.2024.100499] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2023] [Revised: 01/01/2024] [Accepted: 01/02/2024] [Indexed: 01/15/2024] Open
Abstract
Ferroptosis is a novel cell death mechanism that is mediated by iron-dependent lipid peroxidation. It may be involved in atherosclerosis development. Products of phospholipid oxidation play a key role in atherosclerosis. 1-palmitoyl-2-glutaroyl-sn-glycero-3-phosphocholine (PGPC) is a phospholipid oxidation product present in atherosclerotic lesions. It remains unclear whether PGPC causes atherosclerosis by inducing endothelial cell ferroptosis. In this study, human umbilical vein endothelial cells (HUVECs) were treated with PGPC. Intracellular levels of ferrous iron, lipid peroxidation, superoxide anions (O2•-), and glutathione were detected, and expression of fatty acid binding protein-3 (FABP3), glutathione peroxidase 4 (GPX4), and CD36 were measured. Additionally, the mitochondrial membrane potential (MMP) was determined. Aortas from C57BL6 mice were isolated for vasodilation testing. Results showed that PGPC increased ferrous iron levels, the production of lipid peroxidation and O2•-, and FABP3 expression. However, PGPC inhibited the expression of GPX4 and glutathione production and destroyed normal MMP. These effects were also blocked by ferrostatin-1, an inhibitor of ferroptosis. FABP3 silencing significantly reversed the effect of PGPC. Furthermore, PGPC stimulated CD36 expression. Conversely, CD36 silencing reversed the effects of PGPC, including PGPC-induced FABP3 expression. Importantly, E06, a direct inhibitor of the oxidized 1-palmitoyl-2-arachidonoyl-phosphatidylcholine IgM natural antibody, inhibited the effects of PGPC. Finally, PGPC impaired endothelium-dependent vasodilation, ferrostatin-1 or FABP3 inhibitors inhibited this impairment. Our data demonstrate that PGPC impairs endothelial function by inducing endothelial cell ferroptosis through the CD36 receptor to increase FABP3 expression. Our findings provide new insights into the mechanisms of atherosclerosis and a therapeutic target for atherosclerosis.
Collapse
Affiliation(s)
- Si Chen
- Division of Cardiac Surgery, Cardiovascular Diseases Institute, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China; National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, NHC Key Laboratory of Assisted Circulation and Vascular Diseases (Sun Yat-sen University), Key Laboratory of Assisted Circulation and Vascular Diseases, Chinese Academy of Medical Sciences, Guangdong Provincial Engineering and Technology Center for Diagnosis and Treatment of Vascular Diseases, Guangzhou, China
| | - Jian-Jun Gao
- Division of Cardiac Surgery, Cardiovascular Diseases Institute, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China; National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, NHC Key Laboratory of Assisted Circulation and Vascular Diseases (Sun Yat-sen University), Key Laboratory of Assisted Circulation and Vascular Diseases, Chinese Academy of Medical Sciences, Guangdong Provincial Engineering and Technology Center for Diagnosis and Treatment of Vascular Diseases, Guangzhou, China
| | - Yu-Jia Liu
- Division of Cardiac Surgery, Cardiovascular Diseases Institute, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China; National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, NHC Key Laboratory of Assisted Circulation and Vascular Diseases (Sun Yat-sen University), Key Laboratory of Assisted Circulation and Vascular Diseases, Chinese Academy of Medical Sciences, Guangdong Provincial Engineering and Technology Center for Diagnosis and Treatment of Vascular Diseases, Guangzhou, China
| | - Zhi-Wei Mo
- Division of Cardiac Surgery, Cardiovascular Diseases Institute, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China; National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, NHC Key Laboratory of Assisted Circulation and Vascular Diseases (Sun Yat-sen University), Key Laboratory of Assisted Circulation and Vascular Diseases, Chinese Academy of Medical Sciences, Guangdong Provincial Engineering and Technology Center for Diagnosis and Treatment of Vascular Diseases, Guangzhou, China
| | - Fang-Yuan Wu
- National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, NHC Key Laboratory of Assisted Circulation and Vascular Diseases (Sun Yat-sen University), Key Laboratory of Assisted Circulation and Vascular Diseases, Chinese Academy of Medical Sciences, Guangdong Provincial Engineering and Technology Center for Diagnosis and Treatment of Vascular Diseases, Guangzhou, China; Division of Hypertension and Vascular Diseases, Department of Cardiology, Cardiovascular Diseases Institute, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Zuo-Jun Hu
- National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, NHC Key Laboratory of Assisted Circulation and Vascular Diseases (Sun Yat-sen University), Key Laboratory of Assisted Circulation and Vascular Diseases, Chinese Academy of Medical Sciences, Guangdong Provincial Engineering and Technology Center for Diagnosis and Treatment of Vascular Diseases, Guangzhou, China; Division of Vascular Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Yue-Ming Peng
- Division of Cardiac Surgery, Cardiovascular Diseases Institute, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China; National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, NHC Key Laboratory of Assisted Circulation and Vascular Diseases (Sun Yat-sen University), Key Laboratory of Assisted Circulation and Vascular Diseases, Chinese Academy of Medical Sciences, Guangdong Provincial Engineering and Technology Center for Diagnosis and Treatment of Vascular Diseases, Guangzhou, China
| | - Xiao-Qin Zhang
- National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, NHC Key Laboratory of Assisted Circulation and Vascular Diseases (Sun Yat-sen University), Key Laboratory of Assisted Circulation and Vascular Diseases, Chinese Academy of Medical Sciences, Guangdong Provincial Engineering and Technology Center for Diagnosis and Treatment of Vascular Diseases, Guangzhou, China; Division of Hypertension and Vascular Diseases, Department of Cardiology, Cardiovascular Diseases Institute, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Zhen-Sheng Ma
- Division of Cardiac Surgery, Cardiovascular Diseases Institute, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China; National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, NHC Key Laboratory of Assisted Circulation and Vascular Diseases (Sun Yat-sen University), Key Laboratory of Assisted Circulation and Vascular Diseases, Chinese Academy of Medical Sciences, Guangdong Provincial Engineering and Technology Center for Diagnosis and Treatment of Vascular Diseases, Guangzhou, China
| | - Ze-Long Liu
- Division of Cardiac Surgery, Cardiovascular Diseases Institute, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China; National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, NHC Key Laboratory of Assisted Circulation and Vascular Diseases (Sun Yat-sen University), Key Laboratory of Assisted Circulation and Vascular Diseases, Chinese Academy of Medical Sciences, Guangdong Provincial Engineering and Technology Center for Diagnosis and Treatment of Vascular Diseases, Guangzhou, China
| | - Jian-Yun Yan
- Department of Cardiology, Laboratory of Heart Center, Heart Center, Zhujiang Hospital, Southern Medical University, Guangzhou, China; Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, Guangzhou, China; Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular Disease, Guangzhou, China
| | - Zhi-Jun Ou
- National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, NHC Key Laboratory of Assisted Circulation and Vascular Diseases (Sun Yat-sen University), Key Laboratory of Assisted Circulation and Vascular Diseases, Chinese Academy of Medical Sciences, Guangdong Provincial Engineering and Technology Center for Diagnosis and Treatment of Vascular Diseases, Guangzhou, China; Division of Hypertension and Vascular Diseases, Department of Cardiology, Cardiovascular Diseases Institute, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.
| | - Yan Li
- Division of Cardiac Surgery, Cardiovascular Diseases Institute, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China; National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, NHC Key Laboratory of Assisted Circulation and Vascular Diseases (Sun Yat-sen University), Key Laboratory of Assisted Circulation and Vascular Diseases, Chinese Academy of Medical Sciences, Guangdong Provincial Engineering and Technology Center for Diagnosis and Treatment of Vascular Diseases, Guangzhou, China.
| | - Jing-Song Ou
- Division of Cardiac Surgery, Cardiovascular Diseases Institute, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China; National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, NHC Key Laboratory of Assisted Circulation and Vascular Diseases (Sun Yat-sen University), Key Laboratory of Assisted Circulation and Vascular Diseases, Chinese Academy of Medical Sciences, Guangdong Provincial Engineering and Technology Center for Diagnosis and Treatment of Vascular Diseases, Guangzhou, China; Guangdong Provincial Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China.
| |
Collapse
|
5
|
Zhang Y, Wang W, Xu L, Lian Z, Huang J, Chang Y, Guo J, Wang Y, Song K, Ji H. Inflammation-Dependent Association of Lipoprotein (a) with Cardiovascular and Cancer Mortality. Clin Epidemiol 2024; 16:1-4. [PMID: 38222443 PMCID: PMC10787553 DOI: 10.2147/clep.s437456] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Accepted: 12/18/2023] [Indexed: 01/16/2024] Open
Affiliation(s)
- Yiwen Zhang
- Department of Endocrinology, Affiliated Hospital of Qingdao University, Qingdao, People’s Republic of China
| | - Wei Wang
- Department of Hematology, Affiliated Hospital of Qingdao University, Qingdao, People’s Republic of China
| | - Lili Xu
- Department of Endocrinology, Affiliated Hospital of Qingdao University, Qingdao, People’s Republic of China
| | - Zhexun Lian
- Department of Cardiology, Affiliated Hospital of Qingdao University, Qingdao, People’s Republic of China
| | - Jiale Huang
- Department of Cardiology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, People’s Republic of China
| | - Yaowei Chang
- Department of Cardiology, QingPu Branch of Zhongshan Hospital Affiliated to Fudan University, Shanghai, People’s Republic of China
| | - Junjie Guo
- Department of Cardiology, Affiliated Hospital of Qingdao University, Qingdao, People’s Republic of China
| | - Yangang Wang
- Department of Endocrinology, Affiliated Hospital of Qingdao University, Qingdao, People’s Republic of China
| | - Kexiu Song
- Department of International Medicine, Affiliated Hospital of Qingdao University, Qingdao, People’s Republic of China
| | - Hongwei Ji
- Tsinghua Medicine, Tsinghua University, Beijing, People’s Republic of China
- Department of Internal Medicine, Beijing Tsinghua Changgung Hospital, Beijing, People’s Republic of China
| |
Collapse
|
6
|
Bhatia HS, Rikhi R, Allen TS, Yeang C, Guan W, Garg PK, Tsai MY, Criqui MH, Shapiro MD, Tsimikas S. Lipoprotein(a) and the pooled cohort equations for ASCVD risk prediction: The Multi-Ethnic Study of Atherosclerosis. Atherosclerosis 2023; 381:117217. [PMID: 37607461 PMCID: PMC10659123 DOI: 10.1016/j.atherosclerosis.2023.117217] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Revised: 08/02/2023] [Accepted: 08/08/2023] [Indexed: 08/24/2023]
Abstract
BACKGROUND AND AIMS Lipoprotein(a) [Lp(a)] is an independent risk factor for atherosclerotic cardiovascular disease (ASCVD) but is not included in the Pooled Cohort Equations (PCE). We aimed to assess how well the PCE predict 10-year event rates in individuals with elevated Lp(a), and whether the addition of Lp(a) improves risk prediction. METHODS We compared observed versus PCE-predicted 10-year ASCVD event rates, stratified by Lp(a) level and ASCVD risk category using Poisson regression, and evaluated the association between Lp(a) > 50 mg/dL and ASCVD risk using Cox proportional hazards models in the Multi-Ethnic Study of Atherosclerosis (MESA). We evaluated the C-index and net reclassification improvement (NRI) with addition of Lp(a) to the PCE. RESULTS The study population included 6639 individuals (20%, n = 1325 with elevated Lp(a)). The PCE accurately predicted 10-year event rates for individuals with elevated Lp(a) with observed event rates falling within predicted limits. Elevated Lp(a) was associated with increased risk of CVD events overall (HR 1.27, 95% CI 1.00-1.60), particularly in low (HR 2.45, 95% CI 1.40-4.31), and high-risk (HR 1.41, 95% CI 1.02-1.96) individuals. Continuous NRI (95% CI) with the addition of Lp(a) to the PCE for CVD was 0.0963 (0.0158-0.1953) overall, and 0.2999 (0.0876, 0.5525) among low-risk individuals. CONCLUSIONS The PCE performs well for event rate prediction in individuals with elevated Lp(a). However, Lp(a) is associated with increased CVD risk, and the addition of Lp(a) to the PCE improves risk prediction, particularly among low-risk individuals. These results lend support for increasing use of Lp(a) testing for risk assessment.
Collapse
Affiliation(s)
- Harpreet S Bhatia
- Division of Cardiology, Department of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Rishi Rikhi
- Section on Cardiovascular Medicine, Department of Internal Medicine, Wake Forest University School of Medicine, Winston Salem, NC, USA
| | - Tara S Allen
- Division of Preventive Medicine, Department of Family Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Calvin Yeang
- Division of Cardiology, Department of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Weihua Guan
- Division of Biostatistics, School of Public Health, University of Minnesota, Minneapolis, MN, USA
| | - Parveen K Garg
- Division of Cardiology, Department of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Michael Y Tsai
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN, USA
| | - Michael H Criqui
- Division of Cardiology, Department of Medicine, University of California San Diego, La Jolla, CA, USA; Division of Preventive Medicine, Department of Family Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Michael D Shapiro
- Section on Cardiovascular Medicine, Department of Internal Medicine, Wake Forest University School of Medicine, Winston Salem, NC, USA
| | - Sotirios Tsimikas
- Division of Cardiology, Department of Medicine, University of California San Diego, La Jolla, CA, USA.
| |
Collapse
|
7
|
Okobi OE, Odoma VA, Okunromade O, Louise-Oluwasanmi O, Itua B, Ndubuisi C, Ogbeifun OE, Nwatamole BC, Elimihele TA, Adekunle JO, Adekunle AA, Obi CB, Evbayekha EO. Effect of Avocado Consumption on Risk Factors of Cardiovascular Diseases: A Systematic Review and Meta-Analysis. Cureus 2023; 15:e41189. [PMID: 37525782 PMCID: PMC10387226 DOI: 10.7759/cureus.41189] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/30/2023] [Indexed: 08/02/2023] Open
Abstract
High cholesterol levels are a significant risk factor for heart disease, the leading cause of death worldwide. Lowering cholesterol plays a crucial role in maintaining good health. One approach to reducing cholesterol is through dietary modifications, and avocados have been recognized as a potential food choice for this purpose. Avocados are rich in monounsaturated fatty acids (MUFAs), fiber, and plant sterols, which have cholesterol-lowering effects. Incorporating avocados into a low-fat diet can be beneficial. This study design followed the Preferred Reporting Items for Systematic Reviews and Meta-Analysis (PRISMA) guidelines and conducted databases in Cochrane, SCOPUS, PubMed, and Web of Science up until May 2023, combining keywords related to avocados and cardiovascular diseases (CVDs). The study focused on randomized clinical trials (RCTs) and excluded observational studies, meta-analyses, surveys, abstracts, and reviews. Seven RCTs were included in the study, all reporting total cholesterol (TC) levels. The findings of the study showed that individuals who followed an avocado diet experienced reduced TC levels compared to those who followed a habitual diet or a low-fat diet. The avocado group exhibited lower TC levels compared to the control group in both the habitual diet and low-fat diet subgroups. When considering high-density lipoprotein (HDL) levels, the control group had higher HDL levels than the avocado group in the habitual diet subgroup, while the avocado group had higher HDL levels than the control group in the low-fat diet subgroup. In both the habitual diet and low-fat diet subgroups, the avocado group had lower levels of low-density lipoprotein (LDL) compared to the control group. The study concluded that incorporating avocados into the diet can be a beneficial dietary strategy for individuals aiming to lower their cholesterol levels and promote heart health. The avocado diet was associated with decreased LDL levels, but it did not significantly impact triglyceride (TG) levels or fasting glucose levels. Systolic blood pressure values showed minimal changes with the avocado diet.
Collapse
Affiliation(s)
- Okelue E Okobi
- Family Medicine, Larkin Community Hospital, Miami, USA
- Family Medicine, Lakeside Medical Center, Belle Glade, USA
| | - Victor A Odoma
- Cardiology/Oncology, Indiana University (IU) Health Bloomington Hospital, Bloomington, USA
| | - Omolola Okunromade
- Public Health, Jiann-Ping Hsu College of Public Health, Georgia Southern University, Statesboro, USA
| | | | - Blessing Itua
- Internal Medicine, Annotto Bay Hospital, St. Mary, JAM
| | | | | | - Bright C Nwatamole
- Cardiology, Calderdale and Huddersfield NHS Foundation Trust, Huddersfield , GBR
| | - Thomas A Elimihele
- Clinical Research, Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, USA
| | - Joy O Adekunle
- Internal Medicine, Lagos State Health Service (LHSC), Lagos, NGA
| | - Akeem A Adekunle
- Internal Medicine, College of Medicine, University of Lagos, Lagos, NGA
| | | | | |
Collapse
|
8
|
Itabe H, Obama T. The Oxidized Lipoproteins In Vivo: Its Diversity and Behavior in the Human Circulation. Int J Mol Sci 2023; 24:ijms24065747. [PMID: 36982815 PMCID: PMC10053446 DOI: 10.3390/ijms24065747] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2023] [Revised: 03/07/2023] [Accepted: 03/15/2023] [Indexed: 03/19/2023] Open
Abstract
A high concentration of low-density lipoproteins (LDLs) in circulation has been well-known as a major risk factor for cardiovascular diseases. The presence of oxidized LDLs (oxLDLs) in atherosclerotic lesions and circulation was demonstrated using anti-oxLDL monoclonal antibodies. The so-called “oxLDL hypothesis”, as a mechanism for atherosclerosis development, has been attracting attention for decades. However, the oxLDL has been considered a hypothetical particle since the oxLDL present in vivo has not been fully characterized. Several chemically modified LDLs have been proposed to mimic oxLDLs. Some of the subfractions of LDL, especially Lp(a) and electronegative LDL, have been characterized as oxLDL candidates as oxidized phospholipids that stimulate vascular cells. Oxidized high-density lipoprotein (oxHDL) and oxLDL were discovered immunologically in vivo. Recently, an oxLDL-oxHDL complex was found in human plasma, suggesting the involvement of HDLs in the oxidative modification of lipoproteins in vivo. In this review, we summarize our understanding of oxidized lipoproteins and propose a novel standpoint to understand the oxidized lipoproteins present in vivo.
Collapse
|
9
|
Secondary Cardiovascular Prevention after Acute Coronary Syndrome: Emerging Risk Factors and Novel Therapeutic Targets. J Clin Med 2023; 12:jcm12062161. [PMID: 36983163 PMCID: PMC10056379 DOI: 10.3390/jcm12062161] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2023] [Revised: 03/02/2023] [Accepted: 03/07/2023] [Indexed: 03/12/2023] Open
Abstract
The control of cardiovascular risk factors, the promotion of a healthy lifestyle, and antithrombotic therapy are the cornerstones of secondary prevention after acute coronary syndrome (ACS). However, many patients have recurrent ischemic events despite the optimal control of traditional modifiable risk factors and the use of tailored pharmacological therapy, including new-generation antiplatelet and lipid-lowering agents. This evidence emphasizes the importance of identifying novel risk factors and targets to optimize secondary preventive strategies. Lipoprotein(a) (Lp(a)) has emerged as an independent predictor of adverse events after ACS. New molecules such as anti-PCSK9 monoclonal antibodies, small interfering RNAs, and antisense oligonucleotides can reduce plasma Lp(a) levels and are associated with a long-term outcome benefit after the index event. The inflammatory stimulus and the inflammasome, pivotal elements in the development and progression of atherosclerosis, have been widely investigated in patients with coronary artery disease. More recently, randomized clinical trials including post-ACS patients treated with colchicine and monoclonal antibodies targeting cytokines yielded promising results in the reduction in major cardiovascular events after an ACS. Gut dysbiosis has also raised great interest for its potential pathophysiological role in cardiovascular disease. This evidence, albeit preliminary and needing confirmation by larger population-based studies, suggests the possibility of targeting the gut microbiome in particularly high-risk populations. The risk of recurrent ischemic events after ACS is related to the complex interaction between intrinsic predisposing factors and environmental triggers. The identification of novel risk factors and targets is fundamental to customizing patient clinical management with a precision medicine perspective.
Collapse
|
10
|
Clarke R, Hammami I, Sherliker P, Valdes-Marquez E, Watkins H, Hill M, Yang X, Tsimikas S, Hopewell JC. Oxidized phospholipids on apolipoprotein B-100 versus plasminogen and risk of coronary heart disease in the PROCARDIS study. Atherosclerosis 2022; 354:15-22. [PMID: 35803063 DOI: 10.1016/j.atherosclerosis.2022.06.1020] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Revised: 06/16/2022] [Accepted: 06/22/2022] [Indexed: 11/22/2022]
Abstract
BACKGROUND AND AIMS Oxidized phospholipids carried on the apolipoprotein B-100 (OxPL-apoB) component of Lp(a) are predictive of coronary heart disease (CHD), but the role of oxidized phospholipids carried on plasminogen (OxPL-PLG) is unknown. We examined the independent effects of OxPL-apoB and OxPL-PLG for risk of CHD before and after adjustment for Lp(a). METHODS Plasma levels of OxPL-apoB, OxPL-PLG, plasminogen and Lp(a) were measured in the PROCARDIS study of early-onset CHD (906 cases/858 controls). Multivariable logistic regression was used to estimate the odds ratios (OR) for each biomarker with CHD after adjustment for established risk factors. RESULTS Mean levels of OxPL-apoB were higher in cases than controls, but levels of OxPL-PLG and plasminogen were similar. For OxPL-apoB, individuals in the top vs bottom fifth had 2-fold higher age and sex-adjusted OR of CHD (OR = 2.61 [95%CI: 1.91, 3.55]), which were partially attenuated after adjustment for established risk factors. The findings for OxPL-apoB and CHD in PROCARDIS were comparable with those of a meta-analysis of all such studies. However, the associations of OxPL-apoB with CHD were fully attenuated by additional adjustment for Lp(a) (OR = 0.93 [0.54,1.60]). Neither OxPL-PLG nor plasminogen were associated with CHD. Overall, there were no differences in the predictive value for CHD of high vs normal levels (<20th or >80th percentile) of OxPL-apoB, OxPL-PLG, plasminogen or Lp(a) after stratifying for each other. CONCLUSIONS These results highlight the context-dependency of OxPL in plasma and suggest that their associated risk of CHD is chiefly mediated by their carriage on Lp(a).
Collapse
Affiliation(s)
- Robert Clarke
- Clinical Trial Service Unit and Epidemiological Studies Unit, Nuffield Department of Population Health, University of Oxford, Oxford, United Kingdom.
| | - Imen Hammami
- Clinical Trial Service Unit and Epidemiological Studies Unit, Nuffield Department of Population Health, University of Oxford, Oxford, United Kingdom
| | - Paul Sherliker
- Clinical Trial Service Unit and Epidemiological Studies Unit, Nuffield Department of Population Health, University of Oxford, Oxford, United Kingdom
| | - Elsa Valdes-Marquez
- Clinical Trial Service Unit and Epidemiological Studies Unit, Nuffield Department of Population Health, University of Oxford, Oxford, United Kingdom
| | - Hugh Watkins
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Michael Hill
- Clinical Trial Service Unit and Epidemiological Studies Unit, Nuffield Department of Population Health, University of Oxford, Oxford, United Kingdom
| | - Xiaohong Yang
- Division of Cardiovascular Diseases, University of California, San Diego, USA
| | - Sotirios Tsimikas
- Division of Cardiovascular Diseases, University of California, San Diego, USA
| | - Jemma C Hopewell
- Clinical Trial Service Unit and Epidemiological Studies Unit, Nuffield Department of Population Health, University of Oxford, Oxford, United Kingdom.
| | | |
Collapse
|
11
|
Patel N, Mittal N, Choubdar PA, Taub PR. Lipoprotein(a)—When to Screen and How to Treat. CURRENT CARDIOVASCULAR RISK REPORTS 2022. [DOI: 10.1007/s12170-022-00698-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
|
12
|
Schwartz GG, Ballantyne CM. Existing and emerging strategies to lower Lipoprotein(a). Atherosclerosis 2022; 349:110-122. [DOI: 10.1016/j.atherosclerosis.2022.04.020] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/24/2021] [Revised: 04/13/2022] [Accepted: 04/16/2022] [Indexed: 12/24/2022]
|
13
|
Zhang Z, Su Y, Jing R, Qi J, Qi X, Xie Z, Cui B. Acute and lag effects of ambient fine particulate matter on the incidence of dyslipidemia in Chengdu, China: A time-series study. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2022; 29:37919-37929. [PMID: 35072876 DOI: 10.1007/s11356-021-18400-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Accepted: 12/25/2021] [Indexed: 06/14/2023]
Abstract
High levels of ambient fine particulate matter (PM2.5) might increase the risk of death due to cardiovascular diseases (CVDs). As a critical risk factor for CVDs, dyslipidemia can cause CVDs or exacerbate pre-existing ones. This study aimed to investigate whether a short-time exposure to PM2.5 leads to dyslipidemia (HyperTC, HyperLDL-C, HyperTG and HypoHDL-C) in adults. The serum lipid data were provided by the Sichuan Provincial People's Hospital Medical Examination Center. We included 309,654 subjects aged 18-79 between May 10, 2015, and May 10, 2017. An advanced distributed lag nonlinear model (DLNM) was applied to investigate the acute and lag effects of ambient PM2.5 on the risk of dyslipidemia. This study was also stratified by sex, age, BMI and season to examine potential effect modification. We observed that the associations between an interquartile increase in PM2.5 (43 μg/m3) and dyslipidemia were [relative risk (RR); 95% confidence interval (CI)]: 1.042 (1.013, 1.071) for HyperLDL-C and 1.027 (1.006, 1.049) for HyperTC at lag0 day. The lag effects were found at lag6 day for HyperLDL-C, in lag4-6 days for HyperTC and lag4-7 days for HyperTG. Short-term exposure to ambient PM2.5 was related to dyslipidemia and the effect modification was observed in the subgroup analysis. The female and normal-weight populations were more susceptible to the risks of PM2.5 on HyperLDL-C and HyperTC.
Collapse
Affiliation(s)
- Zizheng Zhang
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yang Su
- Clinical Laboratory, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
- Chinese Academy of Sciences Sichuan Translational Medicine Research Hospital, Chengdu, China
| | - Renjie Jing
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jiying Qi
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiaohui Qi
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhen Xie
- Chinese Academy of Sciences Sichuan Translational Medicine Research Hospital, Chengdu, China
- Department of Dermatology, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Bin Cui
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
14
|
Koschinsky ML, Boffa MB. Oxidized phospholipid modification of lipoprotein(a): Epidemiology, biochemistry and pathophysiology. Atherosclerosis 2022; 349:92-100. [DOI: 10.1016/j.atherosclerosis.2022.04.001] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Revised: 03/22/2022] [Accepted: 04/01/2022] [Indexed: 02/05/2023]
|
15
|
Koutsogianni AD, Liberopoulos E, Tellis K, Tselepis AD. Oxidized phospholipids and lipoprotein(a): An update. Eur J Clin Invest 2022; 52:e13710. [PMID: 34837383 DOI: 10.1111/eci.13710] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2021] [Revised: 10/21/2021] [Accepted: 10/21/2021] [Indexed: 12/17/2022]
Abstract
Over the past few years, there has been an undiminished interest in lipoprotein(a) [Lp(a)] and oxidized phospholipids (OxPLs), mainly carried on this lipoprotein. Elevated Lp(a) has been established as an independent causal risk factor for cardiovascular disease. OxPLs play an important role in atherosclerosis. The main questions that remain to be answered, however, is to what extent OxPLs contribute to the atherogenicity of Lp(a), what effect hypolipidaemic medications may have on their levels and the potential clinical benefit of their reduction. This narrative review aimed to summarize currently available data on OxPLs and cardiovascular risk, as well as the effect of established and emerging hypolipidaemic medications on Lp(a)-OxPLs.
Collapse
Affiliation(s)
| | - Evangelos Liberopoulos
- Department of Internal Medicine, Faculty of Medicine, University of Ioannina, Ioannina, Greece
| | - Konstantinos Tellis
- Department of Chemistry, Atherothrombosis Research Centre/Laboratory of Biochemistry, University of Ioannina, Ioannina, Greece
| | - Alexandros D Tselepis
- Department of Chemistry, Atherothrombosis Research Centre/Laboratory of Biochemistry, University of Ioannina, Ioannina, Greece
| |
Collapse
|
16
|
Zhang L, Xue S, Ren F, Huang S, Zhou R, Wang Y, Zhou C, Li Z. An atherosclerotic plaque-targeted single-chain antibody for MR/NIR-II imaging of atherosclerosis and anti-atherosclerosis therapy. J Nanobiotechnology 2021; 19:296. [PMID: 34583680 PMCID: PMC8479957 DOI: 10.1186/s12951-021-01047-4] [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: 07/15/2021] [Accepted: 09/17/2021] [Indexed: 12/11/2022] Open
Abstract
Background Oxidation-specific epitopes (OSEs) are rich in atherosclerotic plaques. Innate and adaptive immune responses to OSEs play an important role in atherosclerosis. The purpose of this study was to develop novel human single-chain variable fragment (scFv) antibody specific to OSEs to image and inhibit atherosclerosis. Results Here, we screened a novel scFv antibody, named as ASA6, from phage-displayed human scFv library. ASA6 can bind to oxidized LDL (Ox-LDL) and atherosclerotic plaques. Meanwhile, ASA6 can also inhibit the uptake of Ox-LDL into macrophage to reduce macrophage apoptosis. The atherosclerotic lesion area of ApoE−/− mice administrated with ASA6 antibody was significantly reduced. Transcriptome analysis reveals the anti-atherosclerosis effect of ASA6 is related to the regulation of fatty acid metabolism and inhibition of M1 macrophage polarization. Moreover, we conjugated ASA6 antibody to NaNdF4@NaGdF4 nanoparticles for noninvasive imaging of atherosclerotic plaques by magnetic resonance (MR) and near-infrared window II (NIR-II) imaging. Conclusions Together, these data demonstrate the potential of ASA6 antibody in targeted therapy and noninvasive imaging for atherosclerosis. Graphic abstract ![]()
Supplementary Information The online version contains supplementary material available at 10.1186/s12951-021-01047-4.
Collapse
Affiliation(s)
- Liwei Zhang
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, 266021, China
| | - Sheng Xue
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, 266021, China.
| | - Feng Ren
- Center for Molecular Imaging and Nuclear Medicine, State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Soochow University, Collaborative Innovation Center of Radiological Medicine of Jiangsu Higher Education Institutions, Suzhou, 215123, China
| | - Siyang Huang
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, 266021, China
| | - Ruizhi Zhou
- Department of Radiology, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, 266021, China
| | - Yu Wang
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, 266021, China
| | - Changyong Zhou
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, 266021, China
| | - Zhen Li
- Center for Molecular Imaging and Nuclear Medicine, State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Soochow University, Collaborative Innovation Center of Radiological Medicine of Jiangsu Higher Education Institutions, Suzhou, 215123, China.
| |
Collapse
|
17
|
Abstract
Lipoprotein(a) [Lp(a)] is an atherogenic lipoprotein with a strong genetic regulation. Up to 90% of the concentrations are explained by a single gene, the LPA gene. The concentrations show a several-hundred-fold interindividual variability ranging from less than 0.1 mg/dL to more than 300 mg/dL. Lp(a) plasma concentrations above 30 mg/dL and even more above 50 mg/dL are associated with an increased risk for cardiovascular disease including myocardial infarction, stroke, aortic valve stenosis, heart failure, peripheral arterial disease, and all-cause mortality. Since concentrations above 50 mg/dL are observed in roughly 20% of the Caucasian population and in an even higher frequency in African-American and Asian-Indian ethnicities, it can be assumed that Lp(a) is one of the most important genetically determined risk factors for cardiovascular disease.Carriers of genetic variants that are associated with high Lp(a) concentrations have a markedly increased risk for cardiovascular events. Studies that used these genetic variants as a genetic instrument to support a causal role for Lp(a) as a cardiovascular risk factor are called Mendelian randomization studies. The principle of this type of studies has been introduced and tested for the first time ever with Lp(a) and its genetic determinants.There are currently no approved pharmacologic therapies that specifically target Lp(a) concentrations. However, some therapies that target primarily LDL cholesterol have also an influence on Lp(a) concentrations. These are mainly PCSK9 inhibitors that lower LDL cholesterol by 60% and Lp(a) by 25-30%. Furthermore, lipoprotein apheresis lowers both, Lp(a) and LDL cholesterol, by about 60-70%. Some sophisticated study designs and statistical analyses provided support that lowering Lp(a) by these therapies also lowers cardiovascular events on top of the effect caused by lowering LDL cholesterol, although this was not the main target of the therapy. Currently, new therapies targeting RNA such as antisense oligonucleotides (ASO) or small interfering RNA (siRNA) against apolipoprotein(a), the main protein of the Lp(a) particle, are under examination and lower Lp(a) concentrations up to 90%. Since these therapies specifically lower Lp(a) concentrations without influencing other lipoproteins, they will serve the last piece of the puzzle whether a decrease of Lp(a) results also in a decrease of cardiovascular events.
Collapse
|
18
|
Moreau PR, Tomas Bosch V, Bouvy-Liivrand M, Õunap K, Örd T, Pulkkinen HH, Pölönen P, Heinäniemi M, Ylä-Herttuala S, Laakkonen JP, Linna-Kuosmanen S, Kaikkonen MU. Profiling of Primary and Mature miRNA Expression in Atherosclerosis-Associated Cell Types. Arterioscler Thromb Vasc Biol 2021; 41:2149-2167. [PMID: 33980036 PMCID: PMC8216629 DOI: 10.1161/atvbaha.121.315579] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Accepted: 04/28/2021] [Indexed: 12/13/2022]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- Pierre R. Moreau
- A. I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio (P.R.M., V.T.B., K.O., T.O., H.H.P., S.Y.-H., J.P.L., S.L.-K., M.U.K.)
| | - Vanesa Tomas Bosch
- A. I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio (P.R.M., V.T.B., K.O., T.O., H.H.P., S.Y.-H., J.P.L., S.L.-K., M.U.K.)
| | - Maria Bouvy-Liivrand
- Institute of Biomedicine, School of Medicine, University of Eastern Finland, Kuopio (M.B.-L., P.P., M.H.)
- Now with Genevia Technologies Oy, Tampere, Finland (M.B.-L.)
| | - Kadri Õunap
- A. I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio (P.R.M., V.T.B., K.O., T.O., H.H.P., S.Y.-H., J.P.L., S.L.-K., M.U.K.)
| | - Tiit Örd
- A. I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio (P.R.M., V.T.B., K.O., T.O., H.H.P., S.Y.-H., J.P.L., S.L.-K., M.U.K.)
| | - Heidi H. Pulkkinen
- A. I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio (P.R.M., V.T.B., K.O., T.O., H.H.P., S.Y.-H., J.P.L., S.L.-K., M.U.K.)
| | - Petri Pölönen
- Institute of Biomedicine, School of Medicine, University of Eastern Finland, Kuopio (M.B.-L., P.P., M.H.)
- Department of Pathology, St. Jude Children’s Research Hospital, Memphis, TN (P.P.)
| | - Merja Heinäniemi
- Institute of Biomedicine, School of Medicine, University of Eastern Finland, Kuopio (M.B.-L., P.P., M.H.)
| | - Seppo Ylä-Herttuala
- A. I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio (P.R.M., V.T.B., K.O., T.O., H.H.P., S.Y.-H., J.P.L., S.L.-K., M.U.K.)
| | - Johanna P. Laakkonen
- A. I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio (P.R.M., V.T.B., K.O., T.O., H.H.P., S.Y.-H., J.P.L., S.L.-K., M.U.K.)
| | - Suvi Linna-Kuosmanen
- A. I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio (P.R.M., V.T.B., K.O., T.O., H.H.P., S.Y.-H., J.P.L., S.L.-K., M.U.K.)
- Now with MIT Computer Science and Artificial Intelligence Laboratory, Cambridge, MA, and Broad Institute of MIT and Harvard, Cambridge, MA (S.L.-K.)
| | - Minna U. Kaikkonen
- A. I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio (P.R.M., V.T.B., K.O., T.O., H.H.P., S.Y.-H., J.P.L., S.L.-K., M.U.K.)
| |
Collapse
|
19
|
Structure and Dynamics of Oxidized Lipoproteins In Vivo: Roles of High-Density Lipoprotein. Biomedicines 2021; 9:biomedicines9060655. [PMID: 34201176 PMCID: PMC8229488 DOI: 10.3390/biomedicines9060655] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Revised: 06/03/2021] [Accepted: 06/04/2021] [Indexed: 01/30/2023] Open
Abstract
Oxidative modification of lipoproteins is implicated in the occurrence and development of atherosclerotic lesions. Earlier studies have elucidated on the mechanisms of foam cell formation and lipid accumulation in these lesions, which is mediated by scavenger receptor-mediated endocytosis of oxidized low-density lipoprotein (oxLDL). Mounting clinical evidence has supported the involvement of oxLDL in cardiovascular diseases. High-density lipoprotein (HDL) is known as anti-atherogenic; however, recent studies have shown circulating oxidized HDL (oxHDL) is related to cardiovascular diseases. A modified structure of oxLDL, which was increased in the plasma of patients with acute myocardial infarction, was characterized. It had two unique features: (1) a fraction of oxLDL accompanied oxHDL, and (2) apoA1 was heavily modified, while modification of apoB, and the accumulation of oxidized phosphatidylcholine (oxPC) and lysophosphatidylcholine (lysoPC) was less pronounced. When LDL and HDL were present at the same time, oxidized lipoproteins actively interacted with each other, and oxPC and lysoPC were transferred to another lipoprotein particle and enzymatically metabolized rapidly. This brief review provides a novel view on the dynamics of oxLDL and oxHDL in circulation.
Collapse
|
20
|
Guo P, He Z, Jalaludin B, Knibbs LD, Leskinen A, Roponen M, Komppula M, Jalava P, Hu L, Chen G, Zeng X, Yang B, Dong G. Short-Term Effects of Particle Size and Constituents on Blood Pressure in Healthy Young Adults in Guangzhou, China. J Am Heart Assoc 2021; 10:e019063. [PMID: 33942624 PMCID: PMC8200702 DOI: 10.1161/jaha.120.019063] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Accepted: 02/04/2021] [Indexed: 12/21/2022]
Abstract
Background Although several studies have focused on the associations between particle size and constituents and blood pressure, results have been inconsistent. Methods and Results We conducted a panel study, between December 2017 and January 2018, in 88 healthy university students in Guangzhou, China. Weekly systolic blood pressure and diastolic blood pressure were measured for each participant for 5 consecutive weeks, resulting in a total of 440 visits. Mass concentrations of particles with an aerodynamic diameter of ≤2.5 µm (PM2.5), ≤1.0 µm (PM1.0), ≤0.5 µm (PM0.5), ≤0.2 µm (PM0.2), and number concentrations of airborne particulates of diameter ≤0.1 μm were measured. Linear mixed-effect models were used to estimate the associations between blood pressure and particles and PM2.5 constituents 0 to 48 hours before blood pressure measurement. PM of all the fractions in the 0.2- to 2.5-μm range were positively associated with systolic blood pressure in the first 24 hours, with the percent changes of effect estimates ranging from 3.5% to 8.8% for an interquartile range increment of PM. PM0.2 was also positively associated with diastolic blood pressure, with an increase of 5.9% (95% CI, 1.0%-11.0%) for an interquartile range increment (5.8 μg/m3) at lag 0 to 24 hours. For PM2.5 constituents, we found positive associations between chloride and diastolic blood pressure (1.7% [95% CI, 0.1%-3.3%]), and negative associations between vanadium and diastolic blood pressure (-1.6% [95% CI, -3.0% to -0.1%]). Conclusions Both particle size and constituent exposure are significantly associated with blood pressure in the first 24 hours following exposure in healthy Chinese adults.
Collapse
Affiliation(s)
- Peng‐Yue Guo
- Guangdong Provincial Engineering Technology Research Center of Environmental Pollution and Health Risk AssessmentDepartment of Occupational and Environmental HealthSchool of Public HealthSun Yat‐sen UniversityGuangzhouChina
| | - Zhi‐Zhou He
- Guangdong Provincial Engineering Technology Research Center of Environmental Pollution and Health Risk AssessmentDepartment of Occupational and Environmental HealthSchool of Public HealthSun Yat‐sen UniversityGuangzhouChina
| | - Bin Jalaludin
- Centre for Air Quality and Health Research and EvaluationGlebeAustralia
- Ingham Institute for Applied Medial ResearchUniversity of New South WalesSydneyAustralia
| | - Luke D. Knibbs
- School of Public HealthThe University of QueenslandHerstonQueenslandAustralia
| | - Ari Leskinen
- Finnish Meteorological InstituteKuopioFinland
- Department of Applied PhysicsUniversity of Eastern FinlandKuopioFinland
| | - Marjut Roponen
- Department of Environmental and Biological SciencesUniversity of Eastern FinlandKuopioFinland
| | | | - Pasi Jalava
- Department of Environmental and Biological SciencesUniversity of Eastern FinlandKuopioFinland
| | - Li‐Wen Hu
- Guangdong Provincial Engineering Technology Research Center of Environmental Pollution and Health Risk AssessmentDepartment of Occupational and Environmental HealthSchool of Public HealthSun Yat‐sen UniversityGuangzhouChina
| | - Gongbo Chen
- Guangdong Provincial Engineering Technology Research Center of Environmental Pollution and Health Risk AssessmentDepartment of Occupational and Environmental HealthSchool of Public HealthSun Yat‐sen UniversityGuangzhouChina
| | - Xiao‐Wen Zeng
- Guangdong Provincial Engineering Technology Research Center of Environmental Pollution and Health Risk AssessmentDepartment of Occupational and Environmental HealthSchool of Public HealthSun Yat‐sen UniversityGuangzhouChina
| | - Bo‐Yi Yang
- Guangdong Provincial Engineering Technology Research Center of Environmental Pollution and Health Risk AssessmentDepartment of Occupational and Environmental HealthSchool of Public HealthSun Yat‐sen UniversityGuangzhouChina
| | - Guang‐Hui Dong
- Guangdong Provincial Engineering Technology Research Center of Environmental Pollution and Health Risk AssessmentDepartment of Occupational and Environmental HealthSchool of Public HealthSun Yat‐sen UniversityGuangzhouChina
| |
Collapse
|
21
|
Shaya GE, Leucker TM, Jones SR, Martin SS, Toth PP. Coronary heart disease risk: Low-density lipoprotein and beyond. Trends Cardiovasc Med 2021; 32:181-194. [PMID: 33872757 DOI: 10.1016/j.tcm.2021.04.002] [Citation(s) in RCA: 62] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/13/2021] [Revised: 03/30/2021] [Accepted: 04/05/2021] [Indexed: 01/06/2023]
Abstract
Coronary heart disease (CHD) is the leading cause of morbidity and mortality world-wide and has been characterized as a chronic immunoinflammatory, fibroproliferative disease fueled by lipids. Great advances have been made in elucidating the complex mechanistic interactions among risk factors associated with CHD, yielding abundant success towards preventive measures and the development of pharmaceuticals to prevent and treat CHD via attenuation of lipoprotein-mediated risk. However, significant residual risk remains. Several potentially modifiable CHD risk factors ostensibly contributing to this residual risk have since come to the fore, including systemic inflammation, diabetes mellitus, high-density lipoprotein, plasma triglycerides (TG) and remnant lipoproteins (RLP), lipoprotein(a) (Lp[a]), and vascular endothelial dysfunction (ED). Herein, we summarize the body of evidence implicating each of these risk factors in residual CHD risk.
Collapse
Affiliation(s)
- Gabriel E Shaya
- Johns Hopkins Ciccarone Center for the Prevention of Cardiovascular Disease, Baltimore, MD, USA
| | - Thorsten M Leucker
- Johns Hopkins Ciccarone Center for the Prevention of Cardiovascular Disease, Baltimore, MD, USA
| | - Steven R Jones
- Johns Hopkins Ciccarone Center for the Prevention of Cardiovascular Disease, Baltimore, MD, USA
| | - Seth S Martin
- Johns Hopkins Ciccarone Center for the Prevention of Cardiovascular Disease, Baltimore, MD, USA
| | - Peter P Toth
- Johns Hopkins Ciccarone Center for the Prevention of Cardiovascular Disease, Baltimore, MD, USA; Community Hospital General Medical Center, Sterling, IL, USA.
| |
Collapse
|
22
|
Ho JH, Adam S, Liu Y, Azmi S, Dhage S, Syed AA, Ammori BJ, Donn R, Heald A, Gibson MJ, Malik RA, Yang X, Durrington PN, Tsimikas S, Soran H. Effect of bariatric surgery on plasma levels of oxidised phospholipids, biomarkers of oxidised LDL and lipoprotein(a). J Clin Lipidol 2021; 15:320-331. [PMID: 33518459 DOI: 10.1016/j.jacl.2020.12.002] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Revised: 12/04/2020] [Accepted: 12/06/2020] [Indexed: 11/26/2022]
Abstract
BACKGROUND Obesity is associated with adverse cardiovascular outcomes and this is improved following bariatric surgery. Oxidised phospholipids (OxPL) are thought to reflect the pro-inflammatory effects of lipoprotein(a) [Lp(a)], and both are independent predictors of cardiovascular disease. OBJECTIVE Our study sought to determine the impact of bariatric surgery on OxPL, biomarkers of oxidised LDL (OxLDL) and Lp(a). METHODS This is a prospective, observational study of 59 patients with severe obesity undergoing bariatric surgery. Blood samples were obtained prior to surgery and at 6 and 12 months after. Sixteen patients attending the tertiary medical weight management clinic at the same centre were also recruited for comparison. Lipid and metabolic blood parameters, OxLDL, OxPL on apolipoprotein B-100 (OxPL-apoB), IgG and IgM autoantibodies to MDA-LDL, IgG and IgM apoB-immune complexes and Lp(a) were measured. RESULTS Reduction in body mass index (BMI) was significant following bariatric surgery, from median 48 kg/m2 at baseline to 37 kg/m2 at 6 months and 33 kg/m2 at 12 months. OxPL-apoB levels decreased significantly at 12 months following surgery [5.0 (3.2-7.4) to 3.8 (3.0-5.5) nM, p = 0.001], while contrastingly, Lp(a) increased significantly [10.2 (3.8-31.9) to 16.9 (4.9-38.6) mg/dl, p = 0.002]. There were significant post-surgical decreases in IgG and IgM biomarkers, particularly at 12 months, while OxLDL remained unchanged. CONCLUSIONS Bariatric surgery results in a significant increase in Lp(a) but reductions in OxPL-apoB and other biomarkers of oxidised lipoproteins, suggesting increased synthetic capacity and reduced oxidative stress. These biomarkers might be clinically useful to monitor physiological effects of weight loss interventions.
Collapse
Affiliation(s)
- Jan Hoong Ho
- Department of Medicine, Manchester University NHS Foundation Trust, Manchester, UK; Lipid Research Group, Division of Medical Sciences, The University of Manchester, Manchester, UK
| | - Safwaan Adam
- Department of Medicine, Manchester University NHS Foundation Trust, Manchester, UK; Lipid Research Group, Division of Medical Sciences, The University of Manchester, Manchester, UK
| | - Yifen Liu
- Lipid Research Group, Division of Medical Sciences, The University of Manchester, Manchester, UK
| | - Shazli Azmi
- Lipid Research Group, Division of Medical Sciences, The University of Manchester, Manchester, UK
| | - Shaishav Dhage
- Department of Medicine, Manchester University NHS Foundation Trust, Manchester, UK; Lipid Research Group, Division of Medical Sciences, The University of Manchester, Manchester, UK
| | - Akheel A Syed
- Department of Diabetes & Endocrinology, Salford Royal NHS Foundation Trust, Salford, UK
| | - Basil J Ammori
- Department of Surgery, Salford Royal NHS Foundation Trust, Salford, UK
| | - Rachelle Donn
- Lipid Research Group, Division of Medical Sciences, The University of Manchester, Manchester, UK
| | - Adrian Heald
- Department of Diabetes & Endocrinology, Salford Royal NHS Foundation Trust, Salford, UK
| | - Martin J Gibson
- Department of Diabetes & Endocrinology, Salford Royal NHS Foundation Trust, Salford, UK
| | - Rayaz A Malik
- Lipid Research Group, Division of Medical Sciences, The University of Manchester, Manchester, UK; Weill Cornell Medicine-Qatar, Doha, Qatar
| | - Xiaohong Yang
- Division of Cardiovascular Medicine, Sulpizio Cardiovascular Center, University of California, San Diego, USA
| | - Paul N Durrington
- Lipid Research Group, Division of Medical Sciences, The University of Manchester, Manchester, UK
| | - Sotirios Tsimikas
- Division of Cardiovascular Medicine, Sulpizio Cardiovascular Center, University of California, San Diego, USA.
| | - Handrean Soran
- Department of Medicine, Manchester University NHS Foundation Trust, Manchester, UK; Lipid Research Group, Division of Medical Sciences, The University of Manchester, Manchester, UK.
| |
Collapse
|
23
|
Khan MI, Hariprasad G. Structural Modeling of Wild and Mutant Forms of Human Plasma Platelet Activating Factor-Acetyl Hydrolase Enzyme. J Inflamm Res 2020; 13:1125-1139. [PMID: 33364808 PMCID: PMC7751442 DOI: 10.2147/jir.s274940] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Accepted: 10/12/2020] [Indexed: 01/09/2023] Open
Abstract
PURPOSE To investigate the structural features of wild and mutant forms of the pPAF-AH enzyme that are responsible for coronary artery disease. METHODS Mutant variants of human pPAF-AH having either V279F, Q281R, or both were modelled and evaluated for stereo chemical and structural correctness. The 3D coordinates of substrate PAF were retrieved from the PubChem database was solvated and minimized on Discovery Studio, and docked to the wild and mutant enzyme models. The top docked pose complex was refined by MD simulation. RESULTS pPAF-AH model comprises of 420 amino acids in a α/β-hydrolase fold that contains a substrate-binding hydrophobic channel with an active site pocket having a catalytic triad of Ser273, Asp296 and His351. Mutations at positions 279 and 281 are opposite one another on the middle of 12 residues long H5 helix that forms the hydrophobic core of the enzyme. V279F causes a tilt on the axis of the mutation bearing helix to avoid steric clashes with the hydrophobic residues on the β-sheets adjacent to it, inducing subtle conformational changes on the H5-β8 loop, β8 sheet, and the loop bearing Asp296. A cascade of conformational changes induces a change in the orientation of His351 resulting in loss of hydrogen bonded interaction with catalytic Ser273. Q281R causes a shortening of H5 and β8, which induces conformational changes of the loops bearing Ser273 and Asp296, respectively. Simultaneous conformational changes of secondary structural elements result in the flipping of His351 causing a break in the catalytic triad. Also, there is a compromise in the substrate-binding area and volume in the mutants resulting in loss of binding to its substrate. CONCLUSION Mutant enzymes show changes at the site of the mutation, secondary motif conformations and global structural conformations that adversely affect the active site, decrease substrate channel volume and decrease stability, thereby affecting enzymatic function.
Collapse
Affiliation(s)
- Mohd Imran Khan
- Department of Biophysics, All India Institute of Medical Sciences, New Delhi110029, India
| | - Gururao Hariprasad
- Department of Biophysics, All India Institute of Medical Sciences, New Delhi110029, India
| |
Collapse
|
24
|
Phosphorylcholine Antibodies Preserve Cardiac Function and Reduce Infarct Size by Attenuating the Post-Ischemic Inflammatory Response. JACC Basic Transl Sci 2020; 5:1228-1239. [PMID: 33426378 PMCID: PMC7775955 DOI: 10.1016/j.jacbts.2020.09.012] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Revised: 09/30/2020] [Accepted: 09/30/2020] [Indexed: 12/12/2022]
Abstract
Phosphorylcholine is a proinflammatory epitope exposed on the outer membrane of apoptotic cells. This study investigated the modulatory effects of a fully human IgG1 monoclonal antibody directed against phosphorylcholine (PC-mAb) on myocardial remodeling and cardiac function following myocardial ischemia-reperfusion injury. PC-mAb attenuates the immediate post-ischemic inflammatory response by reducing the proinflammatory CCL2 chemokine and circulating Ly-6Chi monocytes. This subsequently enhances the post-ischemic repair process resulting in limited adverse cardiac remodeling and preservation of cardiac function. PC-mAb therapy may be a valid therapeutic approach against myocardial ischemia-reperfusion injury.
Phosphorylcholine monoclonal immunoglobulin G antibody attenuates the immediate post-ischemic inflammatory response by reducing the proinflammatory chemokine (C-C motif) ligand 2 chemokine and circulating Ly-6Chi monocytes. This subsequently enhances the post-ischemic repair process, resulting in limited adverse cardiac remodeling and preservation of cardiac function. Therefore, phosphorylcholine monoclonal immunoglobulin G antibody therapy may be a valid therapeutic approach against myocardial ischemia-reperfusion injury.
Collapse
Key Words
- CCL2, chemokine (C-C motif) ligand 2
- CMR, cardiac magnetic resonance
- EDV, end-diastolic volume
- EF, ejection fraction
- ESV, end-systolic volume
- IS, infarct size
- Ig, immunoglobulin
- LV, left ventricular/ventricle
- MI, myocardial infarction
- MI-R, myocardial ischemia-reperfusion
- PC, phosphorylcholine
- PC-mAb, phosphorylcholine monoclonal immunoglobulin G antibody
- cardiac function
- infarct size
- inflammation
- myocardial infarction
- myocardial ischemia-reperfusion
Collapse
|
25
|
Etli M. Investigation of serum ischemia-modified albumin levels in coronary artery disease patients. Indian J Thorac Cardiovasc Surg 2020; 37:147-152. [PMID: 33642712 DOI: 10.1007/s12055-020-01061-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Revised: 09/10/2020] [Accepted: 09/15/2020] [Indexed: 11/28/2022] Open
Abstract
Objective Ischemia-modified albumin (IMA) is a novel marker for the detection of ischemia. The value of this biomarker has been studied in patients with coronary artery disease (CAD). However, the relationship between the severity of coronary stenosis and serum IMA levels remains unknown. Therefore, we aimed to investigate the potential role of serum IMA levels in predicting the severity of coronary atherosclerosis. Materials and methods One hundred and forty-two individuals who underwent coronary angiography for coronary artery disease complaints were included in the study. Participants were divided into three groups based on their diagnosis as control (healthy subjects), group I (subjects with lower Gensini score), and group II (subjects with higher Gensini score). Global Registry of Acute Coronary Events (GRACE) risk score and Gensini scores were calculated after coronary angiogram in the patient groups. Then, venous blood samples were collected from each participant. Serum IMA levels and the levels of routine laboratory markers were measured. Results The serum lymphocyte, neutrophil, and high-density lipid (HDL) levels were statistically insignificant between the groups. The white blood cell (WBC) count and IMA levels were significantly higher in the patient groups (p < 0.05). The GRACE and Gensini scores were significantly different in the patient groups (p < 0.05). However, there was no significant correlation between the GRACE and Gensini scores and serum IMA levels. Conclusion Although IMA levels can be a significant predictor for ischemia according to previous reports, this biomarker seems to be insufficient for determining the severity of disease in patients with CAD.
Collapse
Affiliation(s)
- Mustafa Etli
- Department of Cardiovascular Surgery, Faculty of Medicine, Alanya Alaaddin Keykubat University, Antalya, Turkey
| |
Collapse
|
26
|
Greco MF, Sirtori CR, Corsini A, Ezhov M, Sampietro T, Ruscica M. Lipoprotein(a) Lowering-From Lipoprotein Apheresis to Antisense Oligonucleotide Approach. J Clin Med 2020; 9:jcm9072103. [PMID: 32635396 PMCID: PMC7408876 DOI: 10.3390/jcm9072103] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2020] [Revised: 06/24/2020] [Accepted: 07/01/2020] [Indexed: 12/11/2022] Open
Abstract
It is well-known that elevated lipoprotein(a)—Lp(a)—levels are associated with a higher risk of cardiovascular (CV) mortality and all-cause mortality, although a standard pharmacotherapeutic approach is still undefined for patients with high CV risk dependent on hyperlipoproteinemia(a). Combined with high Lp(a) levels, familial hypercholesterolemia (FH) leads to a greater CVD risk. In suspected FH patients, the proportion of cases explained by a rise of Lp(a) levels ranges between 5% and 20%. In the absence of a specific pharmacological approach able to lower Lp(a) to the extent required to achieve CV benefits, the most effective strategy today is lipoprotein apheresis (LA). Although limited, a clear effect on Lp(a) is exerted by PCSK9 antagonists, with apparently different mechanisms when given with statins (raised catabolism) or as monotherapy (reduced production). In the era of RNA-based therapies, a new dawn is represented by the use of antisense oligonucleotides APO(a)Lrx, able to reduce Lp(a) from 35% to over 80%, with generally modest injection site reactions. The improved knowledge of Lp(a) atherogenicity and possible prevention will be of benefit for patients with residual CV risk remaining after the most effective available lipid-lowering agents.
Collapse
Affiliation(s)
- Maria Francesca Greco
- Dipartimento di Science Farmacologiche e Biomolecolari, Università degli Studi di Milano, 20133 Milan, Italy; (M.F.G.); (A.C.)
| | - Cesare R. Sirtori
- Dyslipidemia Center, A.S.S.T. Grande Ospedale Metropolitano Niguarda, 20162 Milan, Italy;
| | - Alberto Corsini
- Dipartimento di Science Farmacologiche e Biomolecolari, Università degli Studi di Milano, 20133 Milan, Italy; (M.F.G.); (A.C.)
- IRCCS Multimedica, 20099 Milan, Italy
| | - Marat Ezhov
- National Medical Research Center of Cardiology of the Ministry of Health, Moscow, Russia;
| | - Tiziana Sampietro
- U.O. Lipoapheresis and Center for Inherited Dyslipidemias, Fondazione Toscana Gabriele Monasterio, 56126 Pisa, Italy;
| | - Massimiliano Ruscica
- Dipartimento di Science Farmacologiche e Biomolecolari, Università degli Studi di Milano, 20133 Milan, Italy; (M.F.G.); (A.C.)
- Correspondence: ; Tel.: +39-0250318220
| |
Collapse
|
27
|
Ramachandra CJA, Ja KPMM, Chua J, Cong S, Shim W, Hausenloy DJ. Myeloperoxidase As a Multifaceted Target for Cardiovascular Protection. Antioxid Redox Signal 2020; 32:1135-1149. [PMID: 31847538 DOI: 10.1089/ars.2019.7971] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Significance: Myeloperoxidase (MPO) is a heme peroxidase that is primarily expressed by neutrophils. It has the capacity to generate several reactive species, essential for its inherent antimicrobial activity and innate host defense. Dysregulated MPO release, however, can lead to tissue damage, as seen in several diseases. Increased MPO levels in circulation are therefore widely associated with conditions of increased oxidative stress and inflammation. Recent Advances: Several studies have shown a strong correlation between MPO and cardiovascular disease (CVD), through which elevated levels of circulating MPO are linked to poor prognosis with increased risk of CVD-related mortality. Accordingly, circulating MPO is considered a "high-risk" biomarker for patients with acute coronary syndrome, atherosclerosis, heart failure, hypertension, and stroke, thereby implicating MPO as a multifaceted target for cardiovascular protection. Consistently, recent studies that target MPO in animal models of CVD have demonstrated favorable outcomes with regard to disease progression. Critical Issues: Although most of these studies have established a critical link between circulating MPO and worsening cardiac outcomes, the mechanisms by which MPO exerts its detrimental effects in CVD remain unclear. Future Directions: Elucidating the mechanisms by which elevated MPO leads to poor prognosis and, conversely, investigating the beneficial effects of therapeutic MPO inhibition on alleviating disease phenotype will facilitate future MPO-targeted clinical trials for improving CVD-related outcomes.
Collapse
Affiliation(s)
- Chrishan J A Ramachandra
- National Heart Centre Singapore, National Heart Research Institute Singapore, Singapore, Singapore.,Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore Medical School, Singapore, Singapore
| | - K P Myu Mai Ja
- National Heart Centre Singapore, National Heart Research Institute Singapore, Singapore, Singapore
| | - Jasper Chua
- National Heart Centre Singapore, National Heart Research Institute Singapore, Singapore, Singapore.,Faculty of Science, National University of Singapore, Singapore, Singapore
| | - Shuo Cong
- National Heart Centre Singapore, National Heart Research Institute Singapore, Singapore, Singapore.,Department of Cardiac Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Winston Shim
- Health and Social Sciences Cluster, Singapore Institute of Technology, Singapore, Singapore
| | - Derek J Hausenloy
- National Heart Centre Singapore, National Heart Research Institute Singapore, Singapore, Singapore.,Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore Medical School, Singapore, Singapore.,Yong Loo Lin School of Medicine, National University Singapore, Singapore, Singapore.,The Hatter Cardiovascular Institute, University College London, London, United Kingdom.,Cardiovascular Research Center, College of Medical and Health Sciences, Asia University, Taichung, Taiwan
| |
Collapse
|
28
|
Wang L, Tao L, Hao L, Stanley TH, Huang KH, Lambert JD, Kris-Etherton PM. A Moderate-Fat Diet with One Avocado per Day Increases Plasma Antioxidants and Decreases the Oxidation of Small, Dense LDL in Adults with Overweight and Obesity: A Randomized Controlled Trial. J Nutr 2020; 150:276-284. [PMID: 31616932 PMCID: PMC7373821 DOI: 10.1093/jn/nxz231] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2019] [Revised: 05/31/2019] [Accepted: 09/03/2019] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND Avocados are a nutrient-dense source of MUFAs and are rich in antioxidants. Avocados have an additional LDL cholesterol (LDL-C) lowering effect beyond that observed when their MUFAs are substituted for SFAs, especially on small, dense LDL (sdLDL) particles, which are susceptible to in vivo oxidation and associated with increased risk of cardiovascular disease (CVD). OBJECTIVES We investigated whether a healthy diet with 1 avocado daily decreased the following secondary outcomes: circulating oxidized LDL (oxLDL) and related oxidative stress markers. METHODS A randomized, crossover, controlled feeding trial was conducted with 45 men and women, aged 21-70 y, with overweight or obesity and elevated LDL-C (25th-90th percentile). Three cholesterol-lowering diets were provided (5 wk each) in random sequences: a lower-fat (LF) diet (24% calories from fat-7% SFAs, 11% MUFAs, 6% PUFAs) and 2 moderate-fat (MF) diets (34% calories from fat-6% SFAs, 17% MUFAs, 9% PUFAs): the avocado (AV) diet included 1 Hass avocado (∼136 g) per day, and the MF diet used high oleic acid oils to match the fatty acid profile of 1 avocado. A general linear mixed model was used to analyze the treatment effects. RESULTS Compared with baseline, the AV diet significantly decreased circulating oxLDL (-7.0 U/L, -8.8%, P = 0.0004) and increased plasma lutein concentration (19.6 nmol/L, 68.7%, P < 0.0001), and both changes differed significantly from that after the MF and LF diets (P ≤ 0.05). The change in oxLDL caused by the AV diet was significantly correlated with the changes in the number of sdLDL particles (r = 0.32, P = 0.0002) but not large, buoyant LDL particles. CONCLUSIONS One avocado a day in a heart-healthy diet decreased oxLDL in adults with overweight and obesity, and the effect was associated with the reduction in sdLDL. This trial was registered at http://www.clinicaltrials.gov as NCT01235832.
Collapse
Affiliation(s)
- Li Wang
- Department of Nutritional Sciences, Pennsylvania State University, University Park, PA, USA
| | - Ling Tao
- Department of Food Science, Pennsylvania State University, University Park, PA, USA
| | - Lei Hao
- Department of Nutritional Sciences, Pennsylvania State University, University Park, PA, USA
| | - Todd H Stanley
- Department of Food Science, Pennsylvania State University, University Park, PA, USA
| | - Kuan-Hsun Huang
- Department of Nutritional Sciences, Pennsylvania State University, University Park, PA, USA
| | - Joshua D Lambert
- Department of Food Science, Pennsylvania State University, University Park, PA, USA
| | - Penny M Kris-Etherton
- Department of Nutritional Sciences, Pennsylvania State University, University Park, PA, USA
| |
Collapse
|
29
|
Tmoyan NA, Afanasieva OI, Ezhov MV, Klesareva EA, Afanasieva MI, Razova OA, Balakhonova TV, Pokrovsky SN. [Lipoprotein(а) Level, Apolipoprotein(а) Polymorphism аnd Autoаntibodies Against Lipoprotein(а) in Patients with Stenotic Cаrotid Atherosclerosis]. ACTA ACUST UNITED AC 2019; 59:20-27. [PMID: 31849309 DOI: 10.18087/cardio.2019.12.n727] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2019] [Revised: 07/20/2019] [Accepted: 09/17/2019] [Indexed: 11/18/2022]
Abstract
Аim. Comparative assessment of respiratory indicators according to multifunctional monitoring (PFM) with the recommended standard for a complete polysomnographic study and an assessment of the effect of blood pressure (BP) measurements in PFM on sleep quality. Triаls on the аssociаtion of Lp(а) and cаrotid аtherosclerosis аre limited. The аim of the study wаs to investigаte the аssociаtion of Lp(а), аpolipoprotein(а) [apo(а)] polymorphism аnd аutoаntibodies to Lp(а) with stenotic (≥50%) cаrotid аtherosclerosis in dependence on CHD presence. Materials and methods. The study included 785 pаtients аt the аge from 21 to 92 with dаtа of instrumentаl exаmination of coronаry, cаrotid аnd lower limbs аrteries. Stenotic cаrotid аtherosclerosis wаs diаgnosed in 447 pаtients who were divided into two groups depending on presence (n=344) or аbsence (n=103) of CHD. The control group comprised of 338 pаtients without stenotic аtherosclerosis of coronаry, cаrotid аnd lower limbs аrteries. In the blood serum of pаtients levels of Lp(а), аutoаntibodies to Lp(а) were determined аnd аlso аpo(а) phenotyping wаs conducted. Results. There were more mаles, higher аverаge аge аnd frequency of hypertension, type 2 diаbetes mellitus, smoking, Lp(а) concentrаtion (mediаn [interquаrtile rаnge]): 30 [11; 63] vs. 14 [5; 30] mg/dl, p<0.01) in the group with stenotic cаrotid аtherosclerosis in compаrison with control group. Besides, Lp(а) level wаs higher in CHD subgroup thаn in pаtients with stenotic cаrotid аtherosclerosis without CHD: 32 [12; 72] vs. 24 [8; 50] mg/dl, respectively, p=0.01. Elevаted (≥30 mg/dl) Lp(а) level, low moleculаr weight аpolipoprotein(а) [(LMW аpo(а)] phenotype were аssociаted with stenotic cаrotid аtherosclerosis (odds rаtio (OR) 2.9; 95% confidence intervаl (CI) 2.1-4.0, p<0.01 аnd OR 2.3; 95% CI 1.6-3.4, p<0.01, respectively). Logistic regression аnаlysis showed independent аssociаtion of elevаted Lp(а) level аnd LMW аpo(а) phenotype with stenotic cаrotid аtherosclerosis both in the presence аnd absence of CHD. The level of IgM аutoаntibodies to Lp(а) wаs higher in control group thаn in pаtients with stenotic cаrotid аtherosclerosis, p=0.02. Conclusion The level of Lp(a) ≥30 mg/dl and low molecular weight phenotype of aprotein(a) are predictors of stenotic atherosclerosis CA, regardless of the presence of coronary heart disease and other risk factors, while a reverse relationship was found between the level of autoantibodies of the IgM class against Lp(a) and the severity of atherosclerosis CA.
Collapse
Affiliation(s)
- N A Tmoyan
- National Medical Research Center for Cardiology
| | | | - M V Ezhov
- National Medical Research Center for Cardiology
| | | | | | - O A Razova
- National Medical Research Center for Cardiology
| | | | | |
Collapse
|
30
|
Lipoprotein(a): Current Evidence for a Physiologic Role and the Effects of Nutraceutical Strategies. Clin Ther 2019; 41:1780-1797. [DOI: 10.1016/j.clinthera.2019.06.002] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2018] [Revised: 04/01/2019] [Accepted: 06/02/2019] [Indexed: 12/24/2022]
|
31
|
Itabe H, Kato R, Sawada N, Obama T, Yamamoto M. The Significance of Oxidized Low-Density Lipoprotein in Body Fluids as a Marker Related to Diseased Conditions. Curr Med Chem 2019. [PMID: 29521196 DOI: 10.2174/0929867325666180307114855] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Oxidatively modified low-density lipoprotein (oxLDL) is known to be involved in various diseases, including cardiovascular diseases. The presence of oxLDL in the human circulatory system and in atherosclerotic lesions has been demonstrated using monoclonal antibodies. Studies have shown the significance of circulating oxLDL in various systemic diseases, including acute myocardial infarction and diabetic mellitus. Several different enzyme-linked immunosorbent assay (ELISA) procedures to measure oxLDL were utilized. Evidence has been accumulating that reveals changes in oxLDL levels under certain pathological conditions. Since oxLDL concentration tends to correlate with low-density lipoprotein (LDL)-cholesterol, the ratio of ox-LDL and LDL rather than oxLDL concentration alone has also been focused. In addition to circulating plasma, LDL and oxLDL are found in gingival crevicular fluid (GCF), where the ratio of oxLDL to LDL in GCF is much higher than in plasma. LDL and oxLDL levels in GCF show an increase in diabetic patients and periodontal patients, suggesting that GCF might be useful in examining systemic conditions. GCF oxLDL increased when the teeth were affected by periodontitis. It is likely that oxLDL levels in plasma and GCF could reflect oxidative stress and transfer efficacy in the circulatory system.
Collapse
Affiliation(s)
- Hiroyuki Itabe
- Division of Biological Chemistry, Department of Molecular Biology, Showa University School of Pharmacy, Tokyo, Japan
| | - Rina Kato
- Division of Biological Chemistry, Department of Molecular Biology, Showa University School of Pharmacy, Tokyo, Japan
| | - Naoko Sawada
- Division of Biological Chemistry, Department of Molecular Biology, Showa University School of Pharmacy, Tokyo, Japan
| | - Takashi Obama
- Division of Biological Chemistry, Department of Molecular Biology, Showa University School of Pharmacy, Tokyo, Japan
| | - Matsuo Yamamoto
- Department of Periodontology, Showa University School of Dentistry, Tokyo, Japan
| |
Collapse
|
32
|
Schnitzler JG, Dallinga-Thie GM, Kroon J. The Role of (Modified) Lipoproteins in Vascular Function: A Duet Between Monocytes and the Endothelium. Curr Med Chem 2019; 26:1594-1609. [PMID: 29546830 DOI: 10.2174/0929867325666180316121015] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2017] [Revised: 12/05/2017] [Accepted: 12/06/2017] [Indexed: 12/24/2022]
Abstract
Over the last century, many studies have demonstrated that low-density lipoprotein (LDL) is a key risk factor of cardiovascular diseases (CVD) related to atherosclerosis. Thus, for these CVD patients, LDL lowering agents are commonly used in the clinic to reduce the risk for CVD. LDL, upon modification, will develop distinct inflammatory and proatherogenic potential, leading to impaired endothelial integrity, influx of immune cells and subsequent increased foam cell formation. LDL can also directly affect peripheral monocyte composition, rendering them in a more favorable position to migrate and accumulate in the subendothelial space. It has become apparent that other lipoprotein particles, such as triglyceride- rich lipoproteins or remnants (TRL) and lipoprotein(a) [Lp(a)] may also impact on atherogenic pathways. Evidence is accumulating that Lp(a) can promote peripheral monocyte activation, eventually leading to increased transmigration through the endothelium. Similarly, remnant cholesterol has been identified to play a key role in endothelial dysfunction and monocyte behavior. In this review, we will discuss recent developments in understanding the role of different lipoproteins in the context of inflammation at both the level of the monocyte and the endothelium.
Collapse
Affiliation(s)
- Johan G Schnitzler
- Department of Vascular Medicine, Academic Medical Center, University of Amsterdam, Amsterdam, Netherlands
| | - Geesje M Dallinga-Thie
- Department of Vascular Medicine, Academic Medical Center, University of Amsterdam, Amsterdam, Netherlands.,Department of Experimental Vascular Medicine, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Jeffrey Kroon
- Department of Vascular Medicine, Academic Medical Center, University of Amsterdam, Amsterdam, Netherlands.,Department of Experimental Vascular Medicine, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
33
|
Rawther T, Tabet F. Biology, pathophysiology and current therapies that affect lipoprotein (a) levels. J Mol Cell Cardiol 2019; 131:1-11. [DOI: 10.1016/j.yjmcc.2019.04.005] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/22/2018] [Revised: 03/22/2019] [Accepted: 04/09/2019] [Indexed: 12/11/2022]
|
34
|
Boffa MB, Koschinsky ML. Oxidized phospholipids as a unifying theory for lipoprotein(a) and cardiovascular disease. Nat Rev Cardiol 2019; 16:305-318. [DOI: 10.1038/s41569-018-0153-2] [Citation(s) in RCA: 114] [Impact Index Per Article: 22.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
|
35
|
Stamenkovic A, Pierce GN, Ravandi A. Oxidized lipids: not just another brick in the wall 1. Can J Physiol Pharmacol 2018; 97:473-485. [PMID: 30444647 DOI: 10.1139/cjpp-2018-0490] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Over the past decade, there has been intense investigation in trying to understand the pathological role that oxidized phospholipids play in cardiovascular disease. Phospholipids are targets for oxidation, particularly during conditions of excess free radical generation. Once oxidized, they acquire novel roles uncharacteristic of their precursors. Oxidized phosphatidylcholines have an important role in multiple physiological and pathophysiological conditions including atherosclerosis, neurodegenerative diseases, lung disease, inflammation, and chronic alcohol consumption. Circulating oxidized phosphatidylcholine may also serve as a clinical biomarker. The focus of this review, therefore, will be to summarize existing evidence that oxidized phosphatidylcholine molecules play an important role in cardiovascular pathology.
Collapse
Affiliation(s)
- Aleksandra Stamenkovic
- a Institute of Cardiovascular Sciences, St. Boniface Hospital Albrechtsen Research Centre, Winnipeg, MB R2H 2A6, Canada.,b Department of Physiology & Pathophysiology, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB R3T 2N6, Canada
| | - Grant N Pierce
- a Institute of Cardiovascular Sciences, St. Boniface Hospital Albrechtsen Research Centre, Winnipeg, MB R2H 2A6, Canada.,b Department of Physiology & Pathophysiology, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB R3T 2N6, Canada
| | - Amir Ravandi
- a Institute of Cardiovascular Sciences, St. Boniface Hospital Albrechtsen Research Centre, Winnipeg, MB R2H 2A6, Canada.,c Interventional Cardiology, Section of Cardiology, Max Rady College of Medicine, University of Manitoba, Winnipeg, MB R3T 2N2, Canada
| |
Collapse
|
36
|
Cho KI, Sakuma I, Sohn IS, Jo SH, Koh KK. Inflammatory and metabolic mechanisms underlying the calcific aortic valve disease. Atherosclerosis 2018; 277:60-65. [PMID: 30173080 DOI: 10.1016/j.atherosclerosis.2018.08.029] [Citation(s) in RCA: 78] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2018] [Revised: 07/04/2018] [Accepted: 08/24/2018] [Indexed: 12/19/2022]
Abstract
Although calcific aortic stenosis is a very common disease with major adverse cardiovascular events and healthcare costs, there are no effective medical interventions to delay or halt its progression. Cardiometabolic risk factors, including smoking and male sex, are linked to aortic stenosis. Emerging studies have identified important regulatory roles for immunological and inflammatory responses, including oxidized lipids, various cytokines, and biomineralization. Recent clinical and experimental studies in atherosclerosis and osteoporosis have demonstrated that oxidative stress and oxidized lipids decrease bone formation in the skeletal system while they increase bone formation in the cardiovascular system. Multidisciplinary factors contribute to vascular calcification, including inflammation and metabolic regulation of osteogenesis in the cardiovascular system via similar signaling pathways as bone formation. Calcific aortic valve disease (CAVD) is no longer considered a simple passive process of calcium deposition that occurs with advanced age. Biomineralization in CAVD is a complex, regulated process that involves valvular, circulating, bone marrow-derived cells, macrophage heterogeneity and genetic factors along with biochemical and mechanical factors. The current review will discuss the recently discovered important role of inflammation, metabolic risk factors, and molecular and cellular mechanisms that promote CAVD, as well as the link between osteogenic signals in the skeletal and cardiovascular systems. This may inform future therapeutic strategies for CAVD progression.
Collapse
Affiliation(s)
- Kyoung Im Cho
- Department of Cardiology, Kosin University Gospel Hospital, Busan, Republic of Korea
| | - Ichiro Sakuma
- Cardiovascular Medicine, Hokko Memorial Clinic, Sapporo, Japan; Health Science University of Hokkaido, Tobetsu, Japan
| | - Il Suk Sohn
- Department of Cardiology, Cardiovascular Center, Kyung Hee University Hospital at Gangdong, Seoul, Republic of Korea
| | - Sang-Ho Jo
- Department of Cardiology, Hanlym University Hospital at Pyungchon, Pyungchon, Republic of Korea
| | - Kwang Kon Koh
- Department of Cardiovascular Medicine, Heart Center, Gachon University Gil Medical Center, Incheon, Republic of Korea; Gachon Cardiovascular Research Institute, Incheon, Republic of Korea.
| |
Collapse
|
37
|
Abstract
PURPOSE OF REVIEW As the incidence of calcific aortic valve stenosis increases with the aging of the population, improved understanding and novel therapies to reduce its progression and need for aortic valve replacement are urgently needed. RECENT FINDINGS Lipoprotein(a) is the only monogenetic risk factor for calcific aortic stenosis. Elevated levels are a strong, causal, independent risk factor, as demonstrated in epidemiological, genome-wide association studies and Mendelian randomization studies. Lipoprotein(a) is the major lipoprotein carrier of oxidized phospholipids, which are proinflammatory and promote calcification of vascular cells, two key pathophysiological drivers of aortic stenosis. Elevated plasma lipoprotein(a) and oxidized phospholipids predict progression of pre-existing aortic stenosis and need for aortic valve replacement. The failure of statin trials in pre-existing aortic stenosis may be partially due to an increase in lipoprotein(a) and oxidized phospholipid levels caused by statins. Antisense oligonucleotides targeted to apo(a) are in Phase 2 clinical development and shown to lower both lipoprotein(a) and oxidized phospholipids. SUMMARY Lipoprotein(a) and oxidized phospholipids are key therapeutic targets in calcific aortic stenosis. Strategies aimed at potent lipoprotein(a) lowering to normalize levels and/or to suppress the proinflammatory effects of oxidized phospholipids may prevent progression of this disease.
Collapse
|
38
|
Solati Z, Edel AL, Shang Y, O K, Ravandi A. Oxidized phosphatidylcholines are produced in renal ischemia reperfusion injury. PLoS One 2018; 13:e0195172. [PMID: 29684044 PMCID: PMC5912739 DOI: 10.1371/journal.pone.0195172] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2017] [Accepted: 03/16/2018] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND The aim of this study was to determine the individual oxidized phosphatidylcholine (OxPC) molecules generated during renal ischemia/ reperfusion (I/R) injury. METHODS Kidney ischemia was induced in male Sprague-Dawley rats by clamping the left renal pedicle for 45 min followed by reperfusion for either 6h or 24h. Kidney tissue was subjected to lipid extraction. Phospholipids and OxPC species were identified and quantitated using liquid chromatography coupled to electrospray ionization tandem mass spectrometry using internal standards. RESULT We identified fifty-five distinct OxPC in rat kidney following I/R injury. These included a variety of fragmented (aldehyde and carboxylic acid containing species) and non-fragmented products. 1-stearoyl-2-linoleoyl-phosphatidylcholine (SLPC-OH), which is a non-fragmented OxPC and 1-palmitoyl-2-azelaoyl-sn-glycero-3-phosphocholine (PAzPC), which is a fragmented OxPC, were the most abundant OxPC species after 6h and 24 h I/R respectively. Total fragmented aldehyde OxPC were significantly higher in 6h and 24h I/R groups compared to sham operated groups (P = 0.03, 0.001 respectively). Moreover, levels of aldehyde OxPC at 24h I/R were significantly greater than those in 6h I/R (P = 0.007). Fragmented carboxylic acid increased significantly in 24h I/R group compared with sham and 6h I/R groups (P = 0.001, 0.001). Moreover, levels of fragmented OxPC were significantly correlated with creatinine levels (r = 0.885, P = 0.001). Among non-fragmented OxPC, only isoprostanes were elevated significantly in 6h I/R group compared with sham group but not in 24h I/R group (P = 0.01). No significant changes were observed in other non-fragmented OxPC including long chain products and terminal furans. CONCLUSION We have shown for the first time that bioactive OxPC species are produced in renal I/R and their levels increase with increasing time of reperfusion in a kidney model of I/R and correlate with severity of I/R injury. Given the pathological activity of fragmented OxPCs, therapies focused on their reduction may be a mechanism to attenuate renal I/R injury.
Collapse
Affiliation(s)
- Zahra Solati
- Institute of Cardiovascular Sciences, St. Boniface Hospital Research Centre, University of Manitoba, Winnipeg, Manitoba, Canada
- Department of Physiology and Pathophysiology, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Andrea L. Edel
- Institute of Cardiovascular Sciences, St. Boniface Hospital Research Centre, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Yue Shang
- Department of Physiology and Pathophysiology, University of Manitoba, Winnipeg, Manitoba, Canada
- Department of Animal Science, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Karmin O
- Department of Physiology and Pathophysiology, University of Manitoba, Winnipeg, Manitoba, Canada
- Department of Animal Science, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Amir Ravandi
- Institute of Cardiovascular Sciences, St. Boniface Hospital Research Centre, University of Manitoba, Winnipeg, Manitoba, Canada
- Department of Physiology and Pathophysiology, University of Manitoba, Winnipeg, Manitoba, Canada
- * E-mail:
| |
Collapse
|
39
|
Ellis KL, Boffa MB, Sahebkar A, Koschinsky ML, Watts GF. The renaissance of lipoprotein(a): Brave new world for preventive cardiology? Prog Lipid Res 2017; 68:57-82. [DOI: 10.1016/j.plipres.2017.09.001] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2017] [Revised: 09/01/2017] [Accepted: 09/05/2017] [Indexed: 12/24/2022]
|
40
|
Lp(a) in Childhood. CURRENT CARDIOVASCULAR RISK REPORTS 2017. [DOI: 10.1007/s12170-017-0553-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
|
41
|
Gao S, Zhao D, Wang M, Zhao F, Han X, Qi Y, Liu J. Association Between Circulating Oxidized LDL and Atherosclerotic Cardiovascular Disease: A Meta-analysis of Observational Studies. Can J Cardiol 2017; 33:1624-1632. [PMID: 29173602 DOI: 10.1016/j.cjca.2017.07.015] [Citation(s) in RCA: 77] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2017] [Revised: 07/10/2017] [Accepted: 07/20/2017] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Although basic research has suggested that oxidized low-density lipoprotein (ox-LDL) is involved in the pathogenesis of atherosclerosis, population observational studies have yielded conflicting results about the association between circulating ox-LDL and atherosclerotic cardiovascular disease (ASCVD). Therefore, we performed a systematic review and meta-analysis of currently available observational studies to verify the association between circulating ox-LDL and ASCVD. METHODS We systematically searched PubMed and the Cochrane Library from their inception to March 27, 2017, for nested case-control studies, case-cohort studies, and prospective cohort studies on the relationship between ox-LDL and ASCVD. Studies that did not assess the hazard ratio, relative risk, or odds ratio of ox-LDL or did not adjust for other risk factors, or those without examination of ox-LDL before collection of ASCVD occurrences were excluded. The summarized effect size was combined using fixed effect models. Subgroup analyses were performed on the basis of study quality, study design, definition of ASCVD events, effect size types, types of ox-LDL assay, ox-LDL contrast level, and whether low-density lipoprotein cholesterol was adjusted in a multivariate model. RESULTS A total of 12 included studies consisted of 3 nested case-control studies, 1 case-cohort study, 5 hospital-based cohort studies, and 3 community-based cohort studies. The summary effect size of increased circulating ox-LDL was 1.79 (95% confidence interval, 1.56-2.05) for ASCVD. Similar associations were shown in all subgroups. CONCLUSIONS Our findings indicate that increased levels of circulating ox-LDL are associated with clinical ASCVD events. Further well designed community-based cohort studies or intervention studies are needed to confirm our findings.
Collapse
Affiliation(s)
- Shen Gao
- Department of Epidemiology, Beijing An Zhen Hospital, Capital Medical University, Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing, China
| | - Dong Zhao
- Department of Epidemiology, Beijing An Zhen Hospital, Capital Medical University, Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing, China
| | - Miao Wang
- Department of Epidemiology, Beijing An Zhen Hospital, Capital Medical University, Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing, China
| | - Fan Zhao
- Department of Epidemiology, Beijing An Zhen Hospital, Capital Medical University, Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing, China
| | - Xueyu Han
- Department of Epidemiology, Beijing An Zhen Hospital, Capital Medical University, Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing, China
| | - Yue Qi
- Department of Epidemiology, Beijing An Zhen Hospital, Capital Medical University, Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing, China
| | - Jing Liu
- Department of Epidemiology, Beijing An Zhen Hospital, Capital Medical University, Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing, China.
| |
Collapse
|
42
|
Tsimikas S. A Test in Context: Lipoprotein(a): Diagnosis, Prognosis, Controversies, and Emerging Therapies. J Am Coll Cardiol 2017; 69:692-711. [PMID: 28183512 DOI: 10.1016/j.jacc.2016.11.042] [Citation(s) in RCA: 640] [Impact Index Per Article: 91.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/06/2016] [Revised: 11/10/2016] [Accepted: 11/21/2016] [Indexed: 12/14/2022]
Abstract
Evidence that elevated lipoprotein(a) (Lp[a]) levels contribute to cardiovascular disease (CVD) and calcific aortic valve stenosis (CAVS) is substantial. Development of isoform-independent assays, in concert with genetic, epidemiological, translational, and pathophysiological insights, have established Lp(a) as an independent, genetic, and likely causal risk factor for CVD and CAVS. These observations are consistent across a broad spectrum of patients, risk factors, and concomitant therapies, including patients with low-density lipoprotein cholesterol <70 mg/dl. Statins tend to increase Lp(a) levels, possibly contributing to the "residual risk" noted in outcomes trials and at the bedside. Recently approved proprotein convertase subtilisin/kexin-type 9 inhibitors and mipomersen lower Lp(a) 20% to 30%, and emerging RNA-targeted therapies lower Lp(a) >80%. These approaches will allow testing of the "Lp(a) hypothesis" in clinical trials. This review summarizes the current landscape of Lp(a), discusses controversies, and reviews emerging therapies to reduce plasma Lp(a) levels to decrease risk of CVD and CAVS.
Collapse
Affiliation(s)
- Sotirios Tsimikas
- Division of Cardiovascular Medicine, Sulpizio Cardiovascular Center, University of California San Diego, La Jolla, California.
| |
Collapse
|
43
|
Hypercholesterolemia: The role of PCSK9. Arch Biochem Biophys 2017; 625-626:39-53. [DOI: 10.1016/j.abb.2017.06.001] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2017] [Revised: 05/29/2017] [Accepted: 06/02/2017] [Indexed: 01/06/2023]
|
44
|
Kamstrup PR, Hung MY, Witztum JL, Tsimikas S, Nordestgaard BG. Oxidized Phospholipids and Risk of Calcific Aortic Valve Disease: The Copenhagen General Population Study. Arterioscler Thromb Vasc Biol 2017; 37:1570-1578. [PMID: 28572160 DOI: 10.1161/atvbaha.116.308761] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2016] [Accepted: 05/17/2017] [Indexed: 12/26/2022]
Abstract
OBJECTIVE Lipoprotein(a) is causally associated with calcific aortic valve disease (CAVD). Lipoprotein(a) carries proinflammatory and procalcific oxidized phospholipids (OxPL). We tested whether the CAVD risk is mediated by the content of OxPL on lipoprotein(a). APPROACH AND RESULTS A case-control study was performed within the Copenhagen General Population Study (n=87 980), including 725 CAVD cases (1977-2013) and 1413 controls free of cardiovascular disease. OxPL carried by apoB (apolipoprotein B-100; OxPL-apoB) or apolipoprotein(a) (OxPL-apo(a)) containing lipoproteins, lipoprotein(a) levels, LPA kringle IV type 2 repeat, and rs10455872 genetic variants were measured. OxPL-apoB and OxPL-apo(a) levels correlated with lipoprotein(a) levels among cases (r=0.75 and r=0.95; both P<0.001) and controls (r=0.65 and r=0.93; both P<0.001). OxPL-apoB levels associated with risk of CAVD with odds ratios of 1.2 (95% confidence interval [CI]:1.0-1.6) for 34th to 66th percentile levels, 1.6 (95% CI, 1.2-2.1) for 67th to 90th percentile levels, 2.0 (95% CI, 1.3-3.0) for 91st to 95th percentile levels, and 3.4 (95% CI, 2.1-5.5) for levels >95th percentile, versus levels <34th percentile (trend, P<0.001). Corresponding odds ratios for OxPL-apo(a) were 1.2 (95% CI, 1.0-1.5), 1.2(95% CI, 0.9-1.6), 2.1(95% CI, 1.4-3.1), and 2.9(95% CI, 1.9-4.5; trend, P<0.001) and were similar for lipoprotein(a). LPA genotypes associated with OxPL-apoB, OxPL-apo(a), and lipoprotein(a) levels and explained 34%, 46%, and 39%, respectively, of the total variation in levels. LPA genotypes associated with risk of CAVD; a doubling in genetically determined OxPL-apoB, OxPL-apo(a), and lipoprotein(a) levels associated with odds ratio of CAVD of 1.18 (95% CI, 1.10-1.27), 1.09 (95% CI, 1.05-1.13), and 1.09 (95% CI, 1.05-1.14), respectively, comparable to the corresponding observational estimates of 1.27 (95% CI, 1.16-1.39), 1.13 (95% CI, 1.08-1.18), and 1.11 (95% CI, 1.06-1.17). CONCLUSIONS OxPL-apoB and OxPL-apo(a) are novel genetic and potentially causal risk factors for CAVD and may explain the association of lipoprotein(a) with CAVD.
Collapse
Affiliation(s)
- Pia R Kamstrup
- From the Department of Clinical Biochemistry (P.R.K., B.G.N.) and the Copenhagen General Population Study (P.R.K., B.G.N.), Herlev and Gentofte Hospital, Copenhagen University Hospital, Denmark; Department of Medicine, University of California San Diego, La Jolla (M.-Y.H., J.L.W., S.T.); Department of Internal Medicine, School of Medicine, College of Medicine (M.-Y.H.) and Division of Cardiology, Department of Internal Medicine, Shuang Ho Hospital (M.-Y.H.), Taipei Medical University, Taiwan; Graduate Institute of Clinical Medical Sciences, Chang Gung University College of Medicine, Taoyuan, Taiwan (M.-Y.H.); and Faculty of Health and Medical Sciences, University of Copenhagen, Denmark (B.G.N.).
| | - Ming-Yow Hung
- From the Department of Clinical Biochemistry (P.R.K., B.G.N.) and the Copenhagen General Population Study (P.R.K., B.G.N.), Herlev and Gentofte Hospital, Copenhagen University Hospital, Denmark; Department of Medicine, University of California San Diego, La Jolla (M.-Y.H., J.L.W., S.T.); Department of Internal Medicine, School of Medicine, College of Medicine (M.-Y.H.) and Division of Cardiology, Department of Internal Medicine, Shuang Ho Hospital (M.-Y.H.), Taipei Medical University, Taiwan; Graduate Institute of Clinical Medical Sciences, Chang Gung University College of Medicine, Taoyuan, Taiwan (M.-Y.H.); and Faculty of Health and Medical Sciences, University of Copenhagen, Denmark (B.G.N.)
| | - Joseph L Witztum
- From the Department of Clinical Biochemistry (P.R.K., B.G.N.) and the Copenhagen General Population Study (P.R.K., B.G.N.), Herlev and Gentofte Hospital, Copenhagen University Hospital, Denmark; Department of Medicine, University of California San Diego, La Jolla (M.-Y.H., J.L.W., S.T.); Department of Internal Medicine, School of Medicine, College of Medicine (M.-Y.H.) and Division of Cardiology, Department of Internal Medicine, Shuang Ho Hospital (M.-Y.H.), Taipei Medical University, Taiwan; Graduate Institute of Clinical Medical Sciences, Chang Gung University College of Medicine, Taoyuan, Taiwan (M.-Y.H.); and Faculty of Health and Medical Sciences, University of Copenhagen, Denmark (B.G.N.)
| | - Sotirios Tsimikas
- From the Department of Clinical Biochemistry (P.R.K., B.G.N.) and the Copenhagen General Population Study (P.R.K., B.G.N.), Herlev and Gentofte Hospital, Copenhagen University Hospital, Denmark; Department of Medicine, University of California San Diego, La Jolla (M.-Y.H., J.L.W., S.T.); Department of Internal Medicine, School of Medicine, College of Medicine (M.-Y.H.) and Division of Cardiology, Department of Internal Medicine, Shuang Ho Hospital (M.-Y.H.), Taipei Medical University, Taiwan; Graduate Institute of Clinical Medical Sciences, Chang Gung University College of Medicine, Taoyuan, Taiwan (M.-Y.H.); and Faculty of Health and Medical Sciences, University of Copenhagen, Denmark (B.G.N.).
| | - Børge G Nordestgaard
- From the Department of Clinical Biochemistry (P.R.K., B.G.N.) and the Copenhagen General Population Study (P.R.K., B.G.N.), Herlev and Gentofte Hospital, Copenhagen University Hospital, Denmark; Department of Medicine, University of California San Diego, La Jolla (M.-Y.H., J.L.W., S.T.); Department of Internal Medicine, School of Medicine, College of Medicine (M.-Y.H.) and Division of Cardiology, Department of Internal Medicine, Shuang Ho Hospital (M.-Y.H.), Taipei Medical University, Taiwan; Graduate Institute of Clinical Medical Sciences, Chang Gung University College of Medicine, Taoyuan, Taiwan (M.-Y.H.); and Faculty of Health and Medical Sciences, University of Copenhagen, Denmark (B.G.N.)
| |
Collapse
|
45
|
Gao S, Liu J. Association between circulating oxidized low-density lipoprotein and atherosclerotic cardiovascular disease. Chronic Dis Transl Med 2017; 3:89-94. [PMID: 29063061 PMCID: PMC5627698 DOI: 10.1016/j.cdtm.2017.02.008] [Citation(s) in RCA: 76] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2016] [Indexed: 02/05/2023] Open
Abstract
Atherosclerosis is a chronic, progressive disease which eventually leads to coronary heart disease (CHD), ischemic stroke and other atherosclerotic cardiovascular disease (ASCVD). Numerous studies have demonstrated an atherogenic role of oxidized low-density lipoprotein (ox-LDL) in the progression of ASCVD. This article briefly reviews the atherogenic mechanism of ox-LDL, the methods of measuring ox-LDL in the circulation, effect of medical therapy and life-style modification on ox-LDL level, and the association between circulating ox-LDL and atherosclerosis, including clinical ASCVD events and subclinical atherosclerosis, in observational studies.
Collapse
Affiliation(s)
- Shen Gao
- Department of Epidemiology, Beijing An Zhen Hospital, Capital Medical University, Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing 100029, China
| | - Jing Liu
- Department of Epidemiology, Beijing An Zhen Hospital, Capital Medical University, Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing 100029, China
| |
Collapse
|
46
|
Kelly E, Hemphill L. Lipoprotein(a): A Lipoprotein Whose Time Has Come. CURRENT TREATMENT OPTIONS IN CARDIOVASCULAR MEDICINE 2017; 19:48. [DOI: 10.1007/s11936-017-0549-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
47
|
Measom ND, Down KD, Hirst DJ, Jamieson C, Manas ES, Patel VK, Somers DO. Investigation of a Bicyclo[1.1.1]pentane as a Phenyl Replacement within an LpPLA 2 Inhibitor. ACS Med Chem Lett 2017; 8:43-48. [PMID: 28105273 DOI: 10.1021/acsmedchemlett.6b00281] [Citation(s) in RCA: 141] [Impact Index Per Article: 20.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2016] [Accepted: 11/15/2016] [Indexed: 02/07/2023] Open
Abstract
We describe the incorporation of a bicyclo[1.1.1]pentane moiety within two known LpPLA2 inhibitors to act as bioisosteric phenyl replacements. An efficient synthesis to the target compounds was enabled with a dichlorocarbene insertion into a bicyclo[1.1.0]butane system being the key transformation. Potency, physicochemical, and X-ray crystallographic data were obtained to compare the known inhibitors to their bioisosteric counterparts, which showed the isostere was well tolerated and positively impacted on the physicochemical profile.
Collapse
Affiliation(s)
- Nicholas D. Measom
- GlaxoSmithKline, Medicines Research Centre, Gunnels Wood Road, Stevenage, Hertfordshire, SG1 2NY, U.K
- Department
of Pure and Applied Chemistry, University of Strathclyde, Thomas
Graham Building, 295 Cathedral Street, Glasgow, G1 1XL, U.K
| | - Kenneth D. Down
- GlaxoSmithKline, Medicines Research Centre, Gunnels Wood Road, Stevenage, Hertfordshire, SG1 2NY, U.K
| | - David J. Hirst
- GlaxoSmithKline, Medicines Research Centre, Gunnels Wood Road, Stevenage, Hertfordshire, SG1 2NY, U.K
| | - Craig Jamieson
- Department
of Pure and Applied Chemistry, University of Strathclyde, Thomas
Graham Building, 295 Cathedral Street, Glasgow, G1 1XL, U.K
| | - Eric S. Manas
- GlaxoSmithKline, 1250 South Collegeville Road, Collegeville, Pennsylvania 19426-0989, United States
| | - Vipulkumar K. Patel
- GlaxoSmithKline, Medicines Research Centre, Gunnels Wood Road, Stevenage, Hertfordshire, SG1 2NY, U.K
| | - Don O. Somers
- GlaxoSmithKline, Medicines Research Centre, Gunnels Wood Road, Stevenage, Hertfordshire, SG1 2NY, U.K
| |
Collapse
|
48
|
Leibundgut G, Lee JH, Strauss BH, Segev A, Tsimikas S. Acute and long-term effect of percutaneous coronary intervention on serially-measured oxidative, inflammatory, and coagulation biomarkers in patients with stable angina. J Thromb Thrombolysis 2016; 41:569-80. [PMID: 26964999 DOI: 10.1007/s11239-016-1351-6] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
To derive insights into the temporal changes in oxidative, inflammatory and coagulation biomarkers in patients with stable angina undergoing percutaneous coronary intervention (PCI). PCI is associated with a variety of biochemical and mechanical stresses to the vessel wall. Oxidized phospholipids are present on plasminogen (OxPL-PLG) and potentiate fibrinolysis in vitro. We recently showed that OxPL-PLG increase following acute myocardial infarction, suggesting that they are involved in atherothrombosis. Plasma samples were collected before, immediately after, 6 and 24 h, 3 and 7 days, and 1, 3, and 6 months after PCI in 125 patients with stable angina undergoing uncomplicated PCI. Plasminogen levels, OxPL-PLG, and an array of 16 oxidative, inflammatory and coagulation biomarkers were measured with established assays. OxPL-PLG and plasminogen declined significantly immediately post-PCI, rebounded to baseline, peaked at 3 days and slowly returned to baseline by 6 months (p < 0.0001 by ANOVA). The temporal trends to maximal peak in biomarkers were as follows: immediately post PCI: OxPL-apoB and lipoprotein (a); Day 1-the inflammatory biomarker IL-6; Day 3-CRP and coagulation biomarkers OxPL-PLG, plasminogen and tissue plasminogen activity; Day 3 to 7-plasminogen activator inhibitor activity, and complement factor H binding to malondialdehyde-LDL and MDA-LDL IgG; Day 7-30 MDA-LDL IgM, CuOxLDL IgM, and ApoB-IC IgM and IgG; >30 days uPA activity, uPA antigen, CuOxLDL IgG and peptide mimotope to MDA-LDL. Most of the biomarkers trended to baseline by 6 months. PCI results in a specific, temporal sequence of changes in plasma biomarkers. These observations provide insights into the effects of iatrogenic barotrauma and plaque disruption during PCI and suggest avenues of investigation to explain complications of PCI and development of targeted therapies to enhance procedural success.
Collapse
Affiliation(s)
- Gregor Leibundgut
- Division of Cardiology, University of Basel, Basel, Switzerland.,Vascular Medicine Program, Department of Medicine, University of California San Diego, 9500 Gilman Drive, La Jolla, CA, 92993-0682, USA
| | - Jun-Hee Lee
- Division of Cardiology, Kang-Dong Sacred Heart Hospital, Hallym University Medical Center, Seoul, Korea
| | - Bradley H Strauss
- Division of Cardiology, St. Michael's Hospital, Toronto, ON, Canada.,Schulich Heart Center, Sunnybrook Health Sciences Center, University of Toronto, Toronto, ON, Canada
| | - Amit Segev
- The Heart Centre, Chaim Sheba Medical Centre, Tel Hashomer, Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Sotirios Tsimikas
- Vascular Medicine Program, Department of Medicine, University of California San Diego, 9500 Gilman Drive, La Jolla, CA, 92993-0682, USA.
| |
Collapse
|
49
|
Teng N, Maghzal GJ, Talib J, Rashid I, Lau AK, Stocker R. The roles of myeloperoxidase in coronary artery disease and its potential implication in plaque rupture. Redox Rep 2016; 22:51-73. [PMID: 27884085 PMCID: PMC6837458 DOI: 10.1080/13510002.2016.1256119] [Citation(s) in RCA: 125] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Atherosclerosis is the main pathophysiological process underlying coronary artery disease (CAD). Acute complications of atherosclerosis, such as myocardial infarction, are caused by the rupture of vulnerable atherosclerotic plaques, which are characterized by thin, highly inflamed, and collagen-poor fibrous caps. Several lines of evidence mechanistically link the heme peroxidase myeloperoxidase (MPO), inflammation as well as acute and chronic manifestations of atherosclerosis. MPO and MPO-derived oxidants have been shown to contribute to the formation of foam cells, endothelial dysfunction and apoptosis, the activation of latent matrix metalloproteinases, and the expression of tissue factor that can promote the development of vulnerable plaque. As such, detection, quantification and imaging of MPO mass and activity have become useful in cardiac risk stratification, both for disease assessment and in the identification of patients at risk of plaque rupture. This review summarizes the current knowledge about the role of MPO in CAD with a focus on its possible roles in plaque rupture and recent advances to quantify and image MPO in plasma and atherosclerotic plaques.
Collapse
Affiliation(s)
- Nathaniel Teng
- a Vascular Biology Division , Victor Chang Cardiac Research Institute , Darlinghurst , New South Wales , Australia.,b Department of Cardiology , Prince of Wales Hospital , Randwick , New South Wales , Australia
| | - Ghassan J Maghzal
- a Vascular Biology Division , Victor Chang Cardiac Research Institute , Darlinghurst , New South Wales , Australia
| | - Jihan Talib
- a Vascular Biology Division , Victor Chang Cardiac Research Institute , Darlinghurst , New South Wales , Australia
| | - Imran Rashid
- a Vascular Biology Division , Victor Chang Cardiac Research Institute , Darlinghurst , New South Wales , Australia
| | - Antony K Lau
- b Department of Cardiology , Prince of Wales Hospital , Randwick , New South Wales , Australia.,c Faculty of Medicine , University of New South Wales , Sydney , New South Wales , Australia
| | - Roland Stocker
- a Vascular Biology Division , Victor Chang Cardiac Research Institute , Darlinghurst , New South Wales , Australia.,d School of Medical Sciences , University of New South Wales , Sydney , New South Wales , Australia
| |
Collapse
|
50
|
Lee SR, Prasad A, Choi YS, Xing C, Clopton P, Witztum JL, Tsimikas S. LPA Gene, Ethnicity, and Cardiovascular Events. Circulation 2016; 135:251-263. [PMID: 27831500 DOI: 10.1161/circulationaha.116.024611] [Citation(s) in RCA: 83] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2016] [Accepted: 10/24/2016] [Indexed: 12/16/2022]
Abstract
BACKGROUND The relationship of LPA single nucleotide polymorphisms (SNPs), apolipoprotein(a) isoforms, and lipoprotein(a) [Lp(a)] levels with major adverse cardiovascular events (MACE) in different ethnic groups is not well known. METHODS LPA SNPs, apolipoprotein(a) isoforms, Lp(a), and oxidized phospholipids on apolipoprotein B-100 (OxPL-apoB) levels were measured in 1792 black, 1030 white, and 597 Hispanic subjects enrolled in the Dallas Heart Study. Their interdependent relationships and prospective association with MACE after median 9.5-year follow-up were determined. RESULTS LPA SNP rs3798220 was most prevalent in Hispanics (42.38%), rs10455872 in whites (14.27%), and rs9457951 in blacks (32.92%). The correlation of each of these SNPs with the major apolipoprotein(a) isoform size was highly variable and in different directions among ethnic groups. In the entire cohort, Cox regression analysis with multivariable adjustment revealed that quartiles 4 of Lp(a) and OxPL-apoB were associated with hazard ratios (95% confidence interval) for time to MACE of 2.35 (1.50-3.69, P<0.001) and 1.89 (1.26-2.84, P=0.003), respectively, versus quartile 1. Addition of the major apolipoprotein(a) isoform and the 3 LPA SNPs to these models attenuated the risk, but significance was maintained for both Lp(a) and OxPL-apoB. Evaluating time to MACE in specific ethnic groups, Lp(a) was a positive predictor and the size of the major apolipoprotein(a) isoform was an inverse predictor in blacks, the size of the major apolipoprotein(a) isoform was an inverse predictor in whites, and OxPL-apoB was a positive predictor in Hispanics. CONCLUSIONS The prevalence and association of LPA SNPs with size of apolipoprotein(a) isoforms, Lp(a), and OxPL-apoB levels are highly variable and ethnicity-specific. The relationship to MACE is best explained by elevated plasma Lp(a) or OxPL-apoB levels, despite significant ethnic differences in LPA genetic markers.
Collapse
Affiliation(s)
- Sang-Rok Lee
- From Division of Cardiovascular Diseases, Sulpizio Cardiovascular Center, Department of Medicine, University of California San Diego, La Jolla (S.-R.L., Y.-S.C., S.T.); Division of Cardiology, Chonbuk National University Hospital and Chonbuk School of Medicine, Jeonju, Korea (S.-R.L.); Division of Cardiology, Department of Medicine, The University of Texas Health Science Center San Antonio (A.P.); Division of Cardiology, Department of Internal Medicine, College of Medicine, The Catholic University of Korea, Seoul (Y.-S.C.); Eugene McDermott Center for Human Growth and Development, University of Texas Southwestern Medical Center at Dallas (C.X.); Veterans Affairs Medical Center, San Diego, CA (P.C.); and Division of Endocrinology and Metabolism, University of California San Diego, La Jolla (J.L.W.)
| | - Anand Prasad
- From Division of Cardiovascular Diseases, Sulpizio Cardiovascular Center, Department of Medicine, University of California San Diego, La Jolla (S.-R.L., Y.-S.C., S.T.); Division of Cardiology, Chonbuk National University Hospital and Chonbuk School of Medicine, Jeonju, Korea (S.-R.L.); Division of Cardiology, Department of Medicine, The University of Texas Health Science Center San Antonio (A.P.); Division of Cardiology, Department of Internal Medicine, College of Medicine, The Catholic University of Korea, Seoul (Y.-S.C.); Eugene McDermott Center for Human Growth and Development, University of Texas Southwestern Medical Center at Dallas (C.X.); Veterans Affairs Medical Center, San Diego, CA (P.C.); and Division of Endocrinology and Metabolism, University of California San Diego, La Jolla (J.L.W.)
| | - Yun-Seok Choi
- From Division of Cardiovascular Diseases, Sulpizio Cardiovascular Center, Department of Medicine, University of California San Diego, La Jolla (S.-R.L., Y.-S.C., S.T.); Division of Cardiology, Chonbuk National University Hospital and Chonbuk School of Medicine, Jeonju, Korea (S.-R.L.); Division of Cardiology, Department of Medicine, The University of Texas Health Science Center San Antonio (A.P.); Division of Cardiology, Department of Internal Medicine, College of Medicine, The Catholic University of Korea, Seoul (Y.-S.C.); Eugene McDermott Center for Human Growth and Development, University of Texas Southwestern Medical Center at Dallas (C.X.); Veterans Affairs Medical Center, San Diego, CA (P.C.); and Division of Endocrinology and Metabolism, University of California San Diego, La Jolla (J.L.W.)
| | - Chao Xing
- From Division of Cardiovascular Diseases, Sulpizio Cardiovascular Center, Department of Medicine, University of California San Diego, La Jolla (S.-R.L., Y.-S.C., S.T.); Division of Cardiology, Chonbuk National University Hospital and Chonbuk School of Medicine, Jeonju, Korea (S.-R.L.); Division of Cardiology, Department of Medicine, The University of Texas Health Science Center San Antonio (A.P.); Division of Cardiology, Department of Internal Medicine, College of Medicine, The Catholic University of Korea, Seoul (Y.-S.C.); Eugene McDermott Center for Human Growth and Development, University of Texas Southwestern Medical Center at Dallas (C.X.); Veterans Affairs Medical Center, San Diego, CA (P.C.); and Division of Endocrinology and Metabolism, University of California San Diego, La Jolla (J.L.W.)
| | - Paul Clopton
- From Division of Cardiovascular Diseases, Sulpizio Cardiovascular Center, Department of Medicine, University of California San Diego, La Jolla (S.-R.L., Y.-S.C., S.T.); Division of Cardiology, Chonbuk National University Hospital and Chonbuk School of Medicine, Jeonju, Korea (S.-R.L.); Division of Cardiology, Department of Medicine, The University of Texas Health Science Center San Antonio (A.P.); Division of Cardiology, Department of Internal Medicine, College of Medicine, The Catholic University of Korea, Seoul (Y.-S.C.); Eugene McDermott Center for Human Growth and Development, University of Texas Southwestern Medical Center at Dallas (C.X.); Veterans Affairs Medical Center, San Diego, CA (P.C.); and Division of Endocrinology and Metabolism, University of California San Diego, La Jolla (J.L.W.)
| | - Joseph L Witztum
- From Division of Cardiovascular Diseases, Sulpizio Cardiovascular Center, Department of Medicine, University of California San Diego, La Jolla (S.-R.L., Y.-S.C., S.T.); Division of Cardiology, Chonbuk National University Hospital and Chonbuk School of Medicine, Jeonju, Korea (S.-R.L.); Division of Cardiology, Department of Medicine, The University of Texas Health Science Center San Antonio (A.P.); Division of Cardiology, Department of Internal Medicine, College of Medicine, The Catholic University of Korea, Seoul (Y.-S.C.); Eugene McDermott Center for Human Growth and Development, University of Texas Southwestern Medical Center at Dallas (C.X.); Veterans Affairs Medical Center, San Diego, CA (P.C.); and Division of Endocrinology and Metabolism, University of California San Diego, La Jolla (J.L.W.)
| | - Sotirios Tsimikas
- From Division of Cardiovascular Diseases, Sulpizio Cardiovascular Center, Department of Medicine, University of California San Diego, La Jolla (S.-R.L., Y.-S.C., S.T.); Division of Cardiology, Chonbuk National University Hospital and Chonbuk School of Medicine, Jeonju, Korea (S.-R.L.); Division of Cardiology, Department of Medicine, The University of Texas Health Science Center San Antonio (A.P.); Division of Cardiology, Department of Internal Medicine, College of Medicine, The Catholic University of Korea, Seoul (Y.-S.C.); Eugene McDermott Center for Human Growth and Development, University of Texas Southwestern Medical Center at Dallas (C.X.); Veterans Affairs Medical Center, San Diego, CA (P.C.); and Division of Endocrinology and Metabolism, University of California San Diego, La Jolla (J.L.W.).
| |
Collapse
|