1
|
He Y, Yang YJ, Wang ZJ, Tang L. Bibliometric analysis of treatment modalities in calcific aortic valve stenosis. Front Pharmacol 2025; 16:1431311. [PMID: 40183078 PMCID: PMC11966050 DOI: 10.3389/fphar.2025.1431311] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2024] [Accepted: 02/04/2025] [Indexed: 04/05/2025] Open
Abstract
Background Calcific aortic valve stenosis (CAVS) is a common cardiovascular condition associated with significant adverse events and high mortality rates. Unfortunately, there are currently no effective pharmacological treatments to halt or prevent its progression. Through our analysis of global trends and treatment strategies, we have identified valuable insights and promising therapeutic possibilities. Additionally, by utilizing bibliometric and visualization techniques, we provide a comprehensive overview of the current research landscape in this field. Method According to our design idea, we used the Web of Science database to select publications on aortic stenosis and related treatments. Through our VOSviewer and CiteSpace analysis, a total of 787 articles have been analyzed by September 2024. We also summarize and explore the most prolific authors, the most prolific countries, and the journals and institutions that publish the most articles. Results A visual analysis of the collected articles reveals that Canada and the United States have the highest publication volumes in this field. Among institutions, Harvard University in the U.S. leads in publication count, followed by Laval University in Canada and the University of California in the U.S. The top three research hotspots are stenosis, calcification, and progression. The journal with the highest number of publications in this area is Frontiers in Cardiovascular Medicine, followed by Catheterization and Cardiovascular Interventions and Arteriosclerosis, Thrombosis and Vascular Biology. Furthermore, research on CAVS treatment spans various directions and focuses, including therapeutic approaches, pathogenesis, and diagnostic methods. Conclusion Research into CAVS treatment has advanced significantly over the years. While interventional and surgical valve replacement remains the mainstay treatments for aortic stenosis, they are insufficient to fully meet the needs of the patient. Emerging priorities now focus on improving diagnostics, exploring innovative therapies, uncovering disease mechanisms, and developing novel drugs. These findings highlight the evolving demands in this field and underscore the need for continued research to address these challenges.
Collapse
Affiliation(s)
| | | | | | - Liang Tang
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| |
Collapse
|
2
|
Ti Y, Xu D, Qin X, Hu Y, Xu Y, Zhao Q, Bu P, Li J. Mendelian randomization analysis does not support a causal influence between lipoprotein(A) and immune-mediated inflammatory diseases. Sci Rep 2025; 15:3834. [PMID: 39885280 PMCID: PMC11782540 DOI: 10.1038/s41598-025-88375-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Accepted: 01/28/2025] [Indexed: 02/01/2025] Open
Abstract
Observational studies have reported an association between lipoprotein(a) (Lp(a)) and immune-mediated inflammatory diseases (IMIDs). This study used Mendelian Randomization (MR) and multivariable MR (MVMR) to explore the causal relationship between lipoprotein(a) [Lp(a)] and immune-mediated inflammatory diseases (IMIDs). We performed a bidirectional two-sample mendelian randomization analyses based on genome-wide association study (GWAS) summary statistics of Lp(a) and nine IMIDs, specifically celiac disease (CeD), Crohn's disease (CD), ulcerative colitis (UC), inflammatory bowel disease (IBD), multiple sclerosis (MS), psoriasis (Pso), rheumatoid arthritis (RA), systemic lupus erythematosus (SLE), type 1 diabetes (T1D), and summary-level data for lipid traits. Furthermore, we performed MVMR to examine the independence of relationship between Lp(a) and IMIDs after controlling other lipid traits, namely high-density lipoprotein cholesterol (HDL-C), low-density lipoprotein cholesterol (LDL-C) and triglycerides (TG). We didn't observe a causal association between Lp(a) and the risk of IMIDs in univariable and multivariable MR analysis, challenging previous observational studies. However, genetically predicted lipid traits HDL-C was associated with increased risk of Type 1 diabetes (T1D). The identification of potential mechanisms underlying the observed associations in observational studies necessitates further investigation.
Collapse
Affiliation(s)
- Yun Ti
- State Key Laboratory for Innovation and Transformation of Luobing Theory, Key Laboratory of Cardiovascular Remodeling and Function Research of MOE, NHC, CAMS and Shandong Province; Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
| | - Dan Xu
- Department of General Practice, Qilu Hospital of Shandong University, Jinan, China
| | - Xiaoning Qin
- State Key Laboratory for Innovation and Transformation of Luobing Theory, Key Laboratory of Cardiovascular Remodeling and Function Research of MOE, NHC, CAMS and Shandong Province; Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
| | - Yang Hu
- State Key Laboratory for Innovation and Transformation of Luobing Theory, Key Laboratory of Cardiovascular Remodeling and Function Research of MOE, NHC, CAMS and Shandong Province; Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
| | - Yuru Xu
- State Key Laboratory for Innovation and Transformation of Luobing Theory, Key Laboratory of Cardiovascular Remodeling and Function Research of MOE, NHC, CAMS and Shandong Province; Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
| | - Qingzhao Zhao
- State Key Laboratory for Innovation and Transformation of Luobing Theory, Key Laboratory of Cardiovascular Remodeling and Function Research of MOE, NHC, CAMS and Shandong Province; Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
| | - Peili Bu
- State Key Laboratory for Innovation and Transformation of Luobing Theory, Key Laboratory of Cardiovascular Remodeling and Function Research of MOE, NHC, CAMS and Shandong Province; Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
| | - Jingyuan Li
- State Key Laboratory for Innovation and Transformation of Luobing Theory, Key Laboratory of Cardiovascular Remodeling and Function Research of MOE, NHC, CAMS and Shandong Province; Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China.
| |
Collapse
|
3
|
Wang L, Ma C, Liu X, Han W. Association Between Lipoprotein(a) and Dilatation of Different Aortic Segments in Hypertensive Patients. Echocardiography 2025; 42:e70061. [PMID: 39739981 DOI: 10.1111/echo.70061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2024] [Revised: 11/19/2024] [Accepted: 12/09/2024] [Indexed: 01/02/2025] Open
Abstract
PURPOSE There are limited reports on the potential link between Lp(a) and ARDM. Thus, we examined the relationship between Lp(a) and ARDM among hypertensive patients. METHODS We used echocardiography to measure ARDM in 513 consecutively hospitalized patients. namely, the aortic valve annulus (Ava), sinuses of Valsalva (SV), sinotubular junction (STJ), and ascending aorta (AA) in 513 consecutive inpatients. We also examined the Lp(a), and other laboratory profiles of all participants. RESULTS Lp(a) exhibited a positive and independent relationship with the SV diameter (coefficient [β] = 0.330, p = 0.002) and STJ (coefficient [β] = 0.253, p = 0.023), regardless of age, sex, height, or other clinical factors among hypertensive, but not nonhypertensive patients. We also demonstrated that a marked rise in Lp(a) levels was independently associated with SV dilatation (SVD) (OR: 1.006, 95% CI: 1.002-1.009, p = 0.002) and AA dilatation (AAD) (OR: 1.006, 95% CI: 1.000-1.011, p = 0.035) in patients with hypertension. In the subgroup analysis, elevated Lp(a) levels were significantly associated with SV dilatation in all subgroups, and with AAD in males and patients aged 65 years or younger (p < 0.05). The restricted cubic spline analysis indicated a linear association between Lp(a) levels and the risk of both SV and AAD (p < 0.05). CONCLUSIONS Herein, we were the first to report that among hypertensive patients, elevated Lp(a) concentrations were intricately linked to the ARDMs at SV and STJ. Moreover, we revealed that the Lp(a) level was a stand-alone indicator of SVD and AAD.
Collapse
Affiliation(s)
- Lin Wang
- Department of Cardiology, Zhejiang Hospital, Hangzhou, Zhejiang, China
| | - Chaoqun Ma
- The Second College of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Xiaowei Liu
- Department of Cardiology, Zhejiang Hospital, Hangzhou, Zhejiang, China
| | - Wei Han
- Department of Cardiology, Zhejiang Hospital, Hangzhou, Zhejiang, China
| |
Collapse
|
4
|
Jokesch P, Holzer L, Jantscher L, Guttzeit S, Übelhart R, Oskolkova O, Bochkov V, Gesslbauer B. Identification of plasma proteins binding oxidized phospholipids using pull-down proteomics and OxLDL masking assay. J Lipid Res 2025; 66:100704. [PMID: 39566852 PMCID: PMC11696850 DOI: 10.1016/j.jlr.2024.100704] [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: 08/28/2024] [Revised: 10/08/2024] [Accepted: 11/05/2024] [Indexed: 11/22/2024] Open
Abstract
Oxidized phospholipids (OxPLs) are increasingly recognized as toxic and proinflammatory mediators, which raises interest in the mechanisms of their detoxification. Circulating OxPLs are bound and neutralized by plasma proteins, including both antibodies and non-immunoglobulin proteins. The latter group of proteins is essentially not investigated because only three OxPC-binding plasma proteins are currently known. The goal of this work was to characterize a broad spectrum of plasma proteins selectively binding OxPLs. Using pull-down-proteomic analysis, we found about 150 non-immunoglobulin proteins preferentially binding oxidized 1-palmitoyl-2-arachidonoyl-sn-glycero-phosphatidylcholine (OxPAPC) as compared to non-oxidized PAPC. To test if candidate proteins indeed can form a barrier isolating OxPLs from recognition by other proteins, we applied an immune masking assay. Oxidized LDL (OxLDL) immobilized in multiwell plates was used as a carrier of OxPLs, while mAbs recognizing OxPC or OxPE were used as "detectors" showing if OxPLs on the surface of OxLDL are physically accessible to external binding partners. Using an orthogonal combination of pull-down and masking assays we confirmed that previously described OxPL-binding proteins (non-fractionated IgM, CFH, and Apo-M) indeed can bind to and mask OxPC and OxPE on liposomes and OxLDL. Furthermore, we identified additional plasma proteins selectively binding and masking OxPC including Apo-D, Apo-H, pulmonary surfactant-associated protein B, and antithrombin-III. We hypothesize that in addition to circulating antibodies, multiple non-immunoglobulin plasma proteins can also bind OxPLs and modulate their recognition by innate and adaptive immunity.
Collapse
Affiliation(s)
- Philipp Jokesch
- Department of Pharmaceutical Chemistry, Institute of Pharmaceutical Sciences, University of Graz, Graz, Austria
| | - Lisa Holzer
- Department of Pharmaceutical Chemistry, Institute of Pharmaceutical Sciences, University of Graz, Graz, Austria
| | - Lydia Jantscher
- Department of Pharmaceutical Chemistry, Institute of Pharmaceutical Sciences, University of Graz, Graz, Austria
| | | | | | - Olga Oskolkova
- Department of Pharmaceutical Chemistry, Institute of Pharmaceutical Sciences, University of Graz, Graz, Austria
| | - Valery Bochkov
- Department of Pharmaceutical Chemistry, Institute of Pharmaceutical Sciences, University of Graz, Graz, Austria; Field of Excellence BioHealth - University of Graz, Graz, Austria.
| | - Bernd Gesslbauer
- Department of Pharmaceutical Chemistry, Institute of Pharmaceutical Sciences, University of Graz, Graz, Austria.
| |
Collapse
|
5
|
Sakwe NI, Vuong NB, Black PJ, Ball DD, Thomas P, Beasley HK, Hinton A, Ochieng J, Sakwe AM. Annexin A6 modulates the secretion of pro-inflammatory cytokines and exosomes via interaction with SNAP23 in triple negative breast cancer cells. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.22.619710. [PMID: 39484394 PMCID: PMC11527025 DOI: 10.1101/2024.10.22.619710] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/03/2024]
Abstract
Pro-inflammatory cytokines are secreted via the classical pathway from secretory vesicles or the non-classical pathway via extracellular vesicles (EVs), that together, play critical roles in triple-negative breast cancer (TNBC) progression. Annexin A6 (AnxA6) is a Ca 2+ -dependent membrane-binding protein that in TNBC is implicated in cell growth and invasiveness. AnxA6 is associated with EVs, but whether it affects the secretion of proinflammatory cytokines and/or EVs remains to be fully elucidated. To assess if AnxA6 influences the secretion of cytokines and extracellular vesicles, we used cytokine arrays to analyze secreted factors in cleared culture supernatants from control AnxA6 expressing and AnxA6 downregulated MDA-MB-468 TNBC cells. This revealed the diminished secretion of monocyte chemoattractant protein 1 (MCP-1/CCL2), interleukin 8 (IL-8), dickkopf1 (DKK1), throbospondin-1 (TSP-1), and osteopontin (OPN) following AnxA6 downregulation. We also show that the secretion of small EVs is strongly reduced in AnxA6 downregulated cells and that upregulation of AnxA6 promoted the secretion of treatment was also associated with increased EVs associated Rab7, cholesterol, and MCP-1 levels. Moreover, cholesterol content in EVs was significantly higher in AnxA6-expressing cells than in AnxA6 downregulated cells and following chronic lapatinib induced upregulation of AnxA6. Mechanistically, we demonstrate that the secretion of MCP-1 and/or EVs is AnxA6 dependent and that this requires the translocation of AnxA6 to cellular membranes and its interaction with SNAP23. AnxA6 neutralizing antibodies strongly diminished the survival of AnxA6 low TNBC cells but had minimal effects on the survival of TNBC cells expressing relatively high levels of the protein. Together, these data suggest that AnxA6 facilitates the secretion of EVs and proinflammatory cytokines that may be critical for TNBC progression.
Collapse
|
6
|
Brennan PG, Mota L, Aridi T, Patel N, Liang P, Ferran C. Advancements in Omics and Breakthrough Gene Therapies: A Glimpse into the Future of Peripheral Artery Disease. Ann Vasc Surg 2024; 107:229-246. [PMID: 38582204 DOI: 10.1016/j.avsg.2024.01.031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2023] [Accepted: 01/01/2024] [Indexed: 04/08/2024]
Abstract
Peripheral artery disease (PAD), a highly prevalent global disease, associates with significant morbidity and mortality in affected patients. Despite progress in endovascular and open revascularization techniques for advanced PAD, these interventions grapple with elevated rates of arterial restenosis and vein graft failure attributed to intimal hyperplasia (IH). Novel multiomics technologies, coupled with sophisticated analyses tools recently powered by advances in artificial intelligence, have enabled the study of atherosclerosis and IH with unprecedented single-cell and spatial precision. Numerous studies have pinpointed gene hubs regulating pivotal atherogenic and atheroprotective signaling pathways as potential therapeutic candidates. Leveraging advancements in viral and nonviral gene therapy (GT) platforms, gene editing technologies, and cutting-edge biomaterial reservoirs for delivery uniquely positions us to develop safe, efficient, and targeted GTs for PAD-related diseases. Gene therapies appear particularly fitting for ex vivo genetic engineering of IH-resistant vein grafts. This manuscript highlights currently available state-of-the-art multiomics approaches, explores promising GT-based candidates, and details GT delivery modalities employed by our laboratory and others to thwart mid-term vein graft failure caused by IH, as well as other PAD-related conditions. The potential clinical translation of these targeted GTs holds the promise to revolutionize PAD treatment, thereby enhancing patients' quality of life and life expectancy.
Collapse
Affiliation(s)
- Phillip G Brennan
- Division of Vascular and Endovascular Surgery, and Center for Vascular Biology Research, Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA
| | - Lucas Mota
- Division of Vascular and Endovascular Surgery, and Center for Vascular Biology Research, Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA
| | - Tarek Aridi
- Division of Vascular and Endovascular Surgery, and Center for Vascular Biology Research, Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA; Department of Medicine, Indiana University School of Medicine, Indianapolis, IN
| | - Nyah Patel
- Division of Vascular and Endovascular Surgery, and Center for Vascular Biology Research, Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA
| | - Patric Liang
- Division of Vascular and Endovascular Surgery, and Center for Vascular Biology Research, Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA
| | - Christiane Ferran
- Division of Vascular and Endovascular Surgery, and Center for Vascular Biology Research, Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA; Division of Nephrology and the Transplant Institute, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA.
| |
Collapse
|
7
|
Qin T, Ma TY, Huang K, Lu SJ, Zhong JH, Li JJ. Lipoprotein (a)-Related Inflammatory Imbalance: A Novel Horizon for the Development of Atherosclerosis. Curr Atheroscler Rep 2024; 26:383-394. [PMID: 38878139 PMCID: PMC11236888 DOI: 10.1007/s11883-024-01215-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/15/2024] [Indexed: 07/11/2024]
Abstract
PURPOSE OF REVIEW The primary objective of this review is to explore the pathophysiological roles and clinical implications of lipoprotein(a) [Lp(a)] in the context of atherosclerotic cardiovascular disease (ASCVD). We seek to understand how Lp(a) contributes to inflammation and arteriosclerosis, aiming to provide new insights into the mechanisms of ASCVD progression. RECENT FINDINGS Recent research highlights Lp(a) as an independent risk factor for ASCVD. Studies show that Lp(a) not only promotes the inflammatory processes but also interacts with various cellular components, leading to endothelial dysfunction and smooth muscle cell proliferation. The dual role of Lp(a) in both instigating and, under certain conditions, mitigating inflammation is particularly noteworthy. This review finds that Lp(a) plays a complex role in the development of ASCVD through its involvement in inflammatory pathways. The interplay between Lp(a) levels and inflammatory responses highlights its potential as a target for therapeutic intervention. These insights could pave the way for novel approaches in managing and preventing ASCVD, urging further investigation into Lp(a) as a therapeutic target.
Collapse
Affiliation(s)
- Ting Qin
- Department of Cardiology, Haikou Affiliated Hospital of Central South University Xiangya School of Medicine, Hainan, 570208, China
| | - Tian-Yi Ma
- Department of Cardiology, Haikou Affiliated Hospital of Central South University Xiangya School of Medicine, Hainan, 570208, China
| | - Kang Huang
- Department of Cardiology, Haikou Affiliated Hospital of Central South University Xiangya School of Medicine, Hainan, 570208, China
| | - Shi-Juan Lu
- Department of Cardiology, Haikou Affiliated Hospital of Central South University Xiangya School of Medicine, Hainan, 570208, China.
| | - Jiang-Hua Zhong
- Department of Cardiology, Haikou Affiliated Hospital of Central South University Xiangya School of Medicine, Hainan, 570208, China.
| | - Jian-Jun Li
- Cadiometabolic Center, State Key Laboratory of Cardiovascular Diseases, Fu Wai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100037, China.
| |
Collapse
|
8
|
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
|
9
|
Patel P, Rai V, Agrawal DK. Role of oncostatin-M in ECM remodeling and plaque vulnerability. Mol Cell Biochem 2023; 478:2451-2460. [PMID: 36856919 PMCID: PMC10579161 DOI: 10.1007/s11010-023-04673-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Accepted: 02/06/2023] [Indexed: 03/02/2023]
Abstract
Atherosclerosis is a multifactorial inflammatory disease characterized by the development of plaque formation leading to occlusion of the vessel and hypoxia of the tissue supplied by the vessel. Chronic inflammation and altered collagen expression render stable plaque to unstable and increase plaque vulnerability. Thinned and weakened fibrous cap results in plaque rupture and formation of thrombosis and emboli formation leading to acute ischemic events such as stroke and myocardial infarction. Inflammatory mediators including TREM-1, TLRs, MMPs, and immune cells play a critical role in plaque vulnerability. Among the other inflammatory mediators, oncostatin-M (OSM), a pro-inflammatory cytokine, play an important role in the development and progression of atherosclerosis, however, the role of OSM in plaque vulnerability and extracellular matrix remodeling (ECM) is not well understood and studied. Since ECM remodeling plays an important role in atherosclerosis and plaque vulnerability, a detailed investigation on the role of OSM in ECM remodeling and plaque vulnerability is critical. This is important because the role of OSM has been discussed in the context of proliferation of vascular smooth muscle cells and regulation of cytokine expression but the role of OSM is scarcely discussed in relation to ECM remodeling and plaque vulnerability. This review focuses on critically discussing the role of OSM in ECM remodeling and plaque vulnerability.
Collapse
Affiliation(s)
- Parth Patel
- Department of Translational Research, College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, 309 E. Second Street, Pomona, CA, 91766-1854, USA
| | - Vikrant Rai
- Department of Translational Research, College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, 309 E. Second Street, Pomona, CA, 91766-1854, USA
| | - Devendra K Agrawal
- Department of Translational Research, College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, 309 E. Second Street, Pomona, CA, 91766-1854, USA.
| |
Collapse
|
10
|
Assini JM, Clark JR, Youssef A, Xing C, Doerfler AM, Park SH, Saxena L, Yaseen AB, Børen J, Gros R, Bao G, Lagor WR, Boffa MB, Koschinsky ML. High levels of lipoprotein(a) in transgenic mice exacerbate atherosclerosis and promote vulnerable plaque features in a sex-specific manner. Atherosclerosis 2023; 384:117150. [PMID: 37290980 DOI: 10.1016/j.atherosclerosis.2023.05.019] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 05/18/2023] [Accepted: 05/23/2023] [Indexed: 06/10/2023]
Abstract
BACKGROUND AND AIMS Despite increased clinical interest in lipoprotein(a) (Lp(a)), many questions remain about the molecular mechanisms by which it contributes to atherosclerotic cardiovascular disease. Existing murine transgenic (Tg) Lp(a) models are limited by low plasma levels of Lp(a) and have not consistently shown a pro-atherosclerotic effect of Lp(a). METHODS We generated Tg mice expressing both human apolipoprotein(a) (apo(a)) and human apoB-100, with pathogenic levels of plasma Lp(a) (range 87-250 mg/dL). Female and male Lp(a) Tg mice (Tg(LPA+/0;APOB+/0)) and human apoB-100-only controls (Tg(APOB+/0)) (n = 10-13/group) were fed a high-fat, high-cholesterol diet for 12 weeks, with Ldlr knocked down using an antisense oligonucleotide. FPLC was used to characterize plasma lipoprotein profiles. Plaque area and necrotic core size were quantified and immunohistochemical assessment of lesions using a variety of cellular and protein markers was performed. RESULTS Male and female Tg(LPA+/0;APOB+/0) and Tg(APOB+/0) mice exhibited proatherogenic lipoprotein profiles with increased cholesterol-rich VLDL and LDL-sized particles and no difference in plasma total cholesterol between genotypes. Complex lesions developed in the aortic sinus of all mice. Plaque area (+22%), necrotic core size (+25%), and calcified area (+65%) were all significantly increased in female Tg(LPA+/0;APOB+/0) mice compared to female Tg(APOB+/0) mice. Immunohistochemistry of lesions demonstrated that apo(a) deposited in a similar pattern as apoB-100 in Tg(LPA+/0;APOB+/0) mice. Furthermore, female Tg(LPA+/0;APOB+/0) mice exhibited less organized collagen deposition as well as 42% higher staining for oxidized phospholipids (OxPL) compared to female Tg(APOB+/0) mice. Tg(LPA+/0;APOB+/0) mice had dramatically higher levels of plasma OxPL-apo(a) and OxPL-apoB compared to Tg(APOB+/0) mice, and female Tg(LPA+/0;APOB+/0) mice had higher plasma levels of the proinflammatory cytokine MCP-1 (+3.1-fold) compared to female Tg(APOB+/0) mice. CONCLUSIONS These data suggest a pro-inflammatory phenotype exhibited by female Tg mice expressing Lp(a) that appears to contribute to the development of more severe lesions with greater vulnerable features.
Collapse
Affiliation(s)
- Julia M Assini
- Department of Biochemistry, Schulich School of Medicine & Dentistry, The University of Western Ontario, London, Ontario, N6A 5B7, Canada; Robarts Research Institute, Schulich School of Medicine & Dentistry, London, Ontario, Canada
| | - Justin R Clark
- Robarts Research Institute, Schulich School of Medicine & Dentistry, London, Ontario, Canada; Department of Physiology and Pharmacology, Schulich School of Medicine & Dentistry, The University of Western Ontario, London, Ontario, N6A 5B7, Canada
| | - Amer Youssef
- Robarts Research Institute, Schulich School of Medicine & Dentistry, London, Ontario, Canada
| | - Chuce Xing
- Robarts Research Institute, Schulich School of Medicine & Dentistry, London, Ontario, Canada
| | - Alexandria M Doerfler
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, TX, USA
| | - So Hyun Park
- Department of Bioengineering, Rice University, Houston, USA
| | - Lavanya Saxena
- Department of Bioengineering, Rice University, Houston, USA
| | - Adam B Yaseen
- Department of Bioengineering, Rice University, Houston, USA
| | - Jan Børen
- Department of Molecular and Clinical Medicine, University of Gothenburg, Gothenburg, Sweden
| | - Robert Gros
- Robarts Research Institute, Schulich School of Medicine & Dentistry, London, Ontario, Canada; Department of Physiology and Pharmacology, Schulich School of Medicine & Dentistry, The University of Western Ontario, London, Ontario, N6A 5B7, Canada
| | - Gang Bao
- Robarts Research Institute, Schulich School of Medicine & Dentistry, London, Ontario, Canada
| | - William R Lagor
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, TX, USA; Department of Bioengineering, Rice University, Houston, USA
| | - Michael B Boffa
- Department of Biochemistry, Schulich School of Medicine & Dentistry, The University of Western Ontario, London, Ontario, N6A 5B7, Canada; Robarts Research Institute, Schulich School of Medicine & Dentistry, London, Ontario, Canada.
| | - Marlys L Koschinsky
- Robarts Research Institute, Schulich School of Medicine & Dentistry, London, Ontario, Canada; Department of Physiology and Pharmacology, Schulich School of Medicine & Dentistry, The University of Western Ontario, London, Ontario, N6A 5B7, Canada.
| |
Collapse
|
11
|
Tselepis AD. Treatment of Lp(a): Is It the Future or Are We Ready Today? Curr Atheroscler Rep 2023; 25:679-689. [PMID: 37668953 PMCID: PMC10564831 DOI: 10.1007/s11883-023-01141-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/15/2023] [Indexed: 09/06/2023]
Abstract
PURPOSE OF REVIEW The goal of this review is to present the pharmacodynamic effectiveness as well as the clinical efficacy and safety of investigational antisense oligonucleotides (ASOs) and small interference RNAs (siRNAs) drugs that specifically target lipoprotein(a) (Lp(a)). The review will discuss whether the existing lipid-lowering therapies are adequate to treat high Lp(a) levels or whether it is necessary to use the emerging new therapeutic approaches which are based on the current RNA technologies. RECENT FINDINGS Lipoprotein(a) (Lp(a)) is a causal risk factor for atherosclerotic cardiovascular disease (ASCVD), independent of other conventional risk factors. High Lp(a) levels are also independently associated with an increased risk of aortic stenosis progression rate. Plasma Lp(a) levels are primarily genetically determined by variation in the LPA gene coding for apo(a). All secondary prevention trials have demonstrated that the existing hypolipidemic therapies are not adequate to reduce Lp(a) levels to such an extent that could lead to a substantial reduction of ASCVD risk. This has led to the development of new drugs that target the mRNA transcript of LPA and efficiently inhibit Lp(a) synthesis leading to potent Lp(a) reduction. These new drugs are the ASO pelacarsen and the siRNAs olpasiran and SLN360. Recent pharmacodynamic studies showed that all these drugs potently reduce Lp(a) up to 98%, in a dose-dependent manner. Ongoing clinical trials will determine the Lp(a)-lowering efficacy, tolerability, and safety of these drugs as well as their potential effectiveness in reducing the ASCVD risk attributed to high plasma Lp(a) levels. We are not ready today to significantly reduce plasma Lp(a). Emerging therapies potently decrease Lp(a) and ongoing clinical trials will determine their effectiveness in reducing ASCVD risk in subjects with high Lp(a) levels.
Collapse
Affiliation(s)
- Alexandros D Tselepis
- Atherothrombosis Research Centre/Laboratory of Biochemistry, Department of Chemistry, University of Ioannina, 45110, Ioannina, Greece.
| |
Collapse
|
12
|
Malick WA, Goonewardena SN, Koenig W, Rosenson RS. Clinical Trial Design for Lipoprotein(a)-Lowering Therapies: JACC Focus Seminar 2/3. J Am Coll Cardiol 2023; 81:1633-1645. [PMID: 37076218 DOI: 10.1016/j.jacc.2023.02.033] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Revised: 02/21/2023] [Accepted: 02/24/2023] [Indexed: 04/21/2023]
Abstract
Lipoprotein(a) [Lp(a)] is a source of residual risk in patients with atherosclerotic cardiovascular disease (ASCVD). Clinical trials of fully human monoclonal antibodies targeting proprotein convertase subtilisin kexin 9 have shown that reductions in Lp(a) concentrations may be a predictor of event reduction with this class of cholesterol-lowering therapy. With the advent of selective therapies targeting Lp(a) such as antisense oligonucleotides, small-interfering RNA-based therapies, and gene editing, lowering of Lp(a) may lead to reduction in ASCVD. The phase 3 Lp(a)HORIZON (Assessing the Impact of Lipoprotein(a) Lowering with TQJ230 on Major Cardiovascular Events in Patients With CVD) outcomes trial is currently testing the effect of pelacarsen, an antisense oligonucleotide, on ASCVD risk. Olpasiran is a small-interfering RNA that is in a phase 3 clinical trial. As these therapies enter clinical trials, challenges in trial design will have to be addressed to optimize patient selection and outcomes.
Collapse
Affiliation(s)
- Waqas A Malick
- The Zena and Michael A. Wiener Cardiovascular Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | | | - Wolfgang Koenig
- Deutsches Herzzentrum Muenchen, Technische Universitat Muenchen, Munich, DZHK (German Centre for Cardiovascular Research), Partner Site Munich Heart Alliance, Munich, Germany; Institute of Epidemiology and Medical Biometry, University of Ulm, Ulm, Germany
| | - Robert S Rosenson
- The Zena and Michael A. Wiener Cardiovascular Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA.
| |
Collapse
|
13
|
Mechanism of oxidized phospholipid-related inflammatory response in vascular ageing. Ageing Res Rev 2023; 86:101888. [PMID: 36806379 DOI: 10.1016/j.arr.2023.101888] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Revised: 02/05/2023] [Accepted: 02/16/2023] [Indexed: 02/20/2023]
Abstract
Vascular ageing is an important factor in the morbidity and mortality of the elderly. Atherosclerosis is a characteristic disease of vascular ageing, which is closely related to the enhancement of vascular inflammation. Phospholipid oxidation products are important factors in inducing cellular inflammation. Through interactions with vascular cells and immune cells, they regulate intracellular signaling pathways, activate the expression of various cytokines, and affect cell behavior, such as metabolic level, proliferation, apoptosis, etc. Intervention in lipid metabolism and anti-inflammation are the two key pathways of drugs for the treatment of atherosclerosis. This review aims to sort out the signaling pathway of oxidized phospholipids-induced inflammatory factors in vascular cells and immune cells and the mechanism leading to changes in cell behavior, and summarize the therapeutic targets in the inflammatory signaling pathway for the development of atherosclerosis drugs.
Collapse
|
14
|
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: 10] [Impact Index Per Article: 5.0] [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
|
15
|
Zhang X, Centurion F, Misra A, Patel S, Gu Z. Molecularly targeted nanomedicine enabled by inorganic nanoparticles for atherosclerosis diagnosis and treatment. Adv Drug Deliv Rev 2023; 194:114709. [PMID: 36690300 DOI: 10.1016/j.addr.2023.114709] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 12/20/2022] [Accepted: 01/17/2023] [Indexed: 01/22/2023]
Abstract
Atherosclerosis, a chronic cardiovascular disease caused by plaque development in arteries, remains a leading cause of morbidity and mortality. Atherosclerotic plaques are characterized by the expression and regulation of key molecules such as cell surface receptors, cytokines, and signaling pathway proteins, potentially facilitating precise diagnosis and treatment on a molecular level by specifically targeting the characteristic molecules. In this review, we highlight the recent progress in the past five years on developing molecularly targeted nanomedicine for imaging detection and treatment of atherosclerosis with the use of inorganic nanoparticles. Through targeted delivery of imaging contrast nanoparticles to specific molecules in atherogenesis, atherosclerotic plaque development at different stages could be identified and monitored via various molecular imaging modalities. We also review molecularly targeted therapeutic approaches that target and regulate molecules associated with lipid regulation, inflammation, and apoptosis. The review is concluded with discussion on current challenges and future development of nanomedicine for atherosclerotic diagnosis and treatment.
Collapse
Affiliation(s)
- Xiuwen Zhang
- School of Chemical Engineering, University of New South Wales, Sydney, NSW 2052, Australia
| | - Franco Centurion
- School of Chemical Engineering, University of New South Wales, Sydney, NSW 2052, Australia
| | - Ashish Misra
- Heart Research Institute, Sydney, NSW 2042, Australia; Faculty of Medicine and Health, The University of Sydney, NSW 2006, Australia
| | - Sanjay Patel
- Heart Research Institute, Sydney, NSW 2042, Australia; Royal Prince Alfred Hospital, Sydney, NSW 2050, Australia; Sydney Medical School, The University of Sydney, NSW 2006, Australia
| | - Zi Gu
- School of Chemical Engineering, University of New South Wales, Sydney, NSW 2052, Australia; Australian Centre for NanoMedicine (ACN), University of New South Wales, Sydney, NSW 2052, Australia; UNSW RNA Institute, University of New South Wales, Sydney, NSW 2052, Australia.
| |
Collapse
|
16
|
Khongrum J, Yingthongchai P, Boonyapranai K, Wongtanasarasin W, Aobchecy P, Tateing S, Prachansuwan A, Sitdhipol J, Niwasabutra K, Thaveethaptaikul P, Phapugrangkul P, Chonpathompikunlert P. Safety and Effects of Lactobacillus paracasei TISTR 2593 Supplementation on Improving Cholesterol Metabolism and Atherosclerosis-Related Parameters in Subjects with Hypercholesterolemia: A Randomized, Double-Blind, Placebo-Controlled Clinical Trial. Nutrients 2023; 15:661. [PMID: 36771367 PMCID: PMC9921995 DOI: 10.3390/nu15030661] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Revised: 01/22/2023] [Accepted: 01/23/2023] [Indexed: 02/03/2023] Open
Abstract
Probiotics have the potential as a multi-target approach to modulate hypercholesterolemia associated with premature atherosclerosis. Various strains of Lactobacillus paracasei have been reported to affect hypercholesterolemia positively. This study aimed to investigate the effects of L. paracasei TISTR 2593 on lipid profile, cholesterol metabolism, and atherosclerosis according to the registration of Thai Clinical Trial Registry as identification number TCTR 20220917002. A total of 50 participants with hypercholesterolemia were randomly and equally assigned to consume L. paracasei TISTR 2593 or a placebo in maltodextrin capsules daily. Biomarkers of lipid profiles, oxidative stress state, inflammatory state, and other biological indicators were examined on days 0, 45, and 90. The results showed that subjects taking the L. paracasei TISTR 2593 could significantly reduce the level of serum low-density lipoprotein-cholesterol (p < 0.05), malondialdehyde (p < 0.001), and tumor necrosis factor-α (p < 0.01). Moreover, L. paracasei TISTR 2593 increased the level of serum apolipoprotein E (p < 0.01) and adiponectin (p < 0.001) significantly. No changes in serum total cholesterol, high-density lipoprotein-cholesterol, triglyceride, total bile acids, and monocyte chemoattractant protein-1 were observed during L. paracasei TISTR 2593 supplementation. Therefore, L. paracasei TISTR 2593 could be an adjuvant probiotic supplement to ameliorate hypercholesterolemia and prevent or delay the development of atherosclerosis.
Collapse
Affiliation(s)
- Jurairat Khongrum
- Science and Technology Research Institute, Chiang Mai University, Chiang Mai 50200, Thailand
- Functional Food Research Center for Well-Being, Science and Technology Research Institute, Chiang Mai University, Chiang Mai 50200, Thailand
| | | | - Kongsak Boonyapranai
- Research Institute for Health Science, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Wachira Wongtanasarasin
- Department of Emergency Medicine, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Paitoon Aobchecy
- Science and Technology Research Institute, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Suriya Tateing
- Department of Plant and Soil Sciences, Faculty of Agriculture, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Aree Prachansuwan
- Institute of Nutrition, Mahidol University, Nakhon Pathom 73170, Thailand
| | - Jaruwan Sitdhipol
- Biodiversity Research Centre (BRC), Thailand Institute of Scientific and Technological Research (TISTR), Pathumthani 12120, Thailand
| | - Kanidta Niwasabutra
- Biodiversity Research Centre (BRC), Thailand Institute of Scientific and Technological Research (TISTR), Pathumthani 12120, Thailand
| | - Punnathon Thaveethaptaikul
- Biodiversity Research Centre (BRC), Thailand Institute of Scientific and Technological Research (TISTR), Pathumthani 12120, Thailand
| | - Pongsathon Phapugrangkul
- Biodiversity Research Centre (BRC), Thailand Institute of Scientific and Technological Research (TISTR), Pathumthani 12120, Thailand
| | - Pennapa Chonpathompikunlert
- Biodiversity Research Centre (BRC), Thailand Institute of Scientific and Technological Research (TISTR), Pathumthani 12120, Thailand
| |
Collapse
|
17
|
Lipoprotein(a) in Atherosclerotic Diseases: From Pathophysiology to Diagnosis and Treatment. Molecules 2023; 28:molecules28030969. [PMID: 36770634 PMCID: PMC9918959 DOI: 10.3390/molecules28030969] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Revised: 01/12/2023] [Accepted: 01/17/2023] [Indexed: 01/21/2023] Open
Abstract
Lipoprotein(a) (Lp(a)) is a low-density lipoprotein (LDL) cholesterol-like particle bound to apolipoprotein(a). Increased Lp(a) levels are an independent, heritable causal risk factor for atherosclerotic cardiovascular disease (ASCVD) as they are largely determined by variations in the Lp(a) gene (LPA) locus encoding apo(a). Lp(a) is the preferential lipoprotein carrier for oxidized phospholipids (OxPL), and its role adversely affects vascular inflammation, atherosclerotic lesions, endothelial function and thrombogenicity, which pathophysiologically leads to cardiovascular (CV) events. Despite this crucial role of Lp(a), its measurement lacks a globally unified method, and, between different laboratories, results need standardization. Standard antilipidemic therapies, such as statins, fibrates and ezetimibe, have a mediocre effect on Lp(a) levels, although it is not yet clear whether such treatments can affect CV events and prognosis. This narrative review aims to summarize knowledge regarding the mechanisms mediating the effect of Lp(a) on inflammation, atherosclerosis and thrombosis and discuss current diagnostic and therapeutic potentials.
Collapse
|
18
|
Simantiris S, Antonopoulos AS, Papastamos C, Benetos G, Koumallos N, Tsioufis K, Tousoulis D. Lipoprotein(a) and inflammation- pathophysiological links and clinical implications for cardiovascular disease. J Clin Lipidol 2023; 17:55-63. [PMID: 36333256 DOI: 10.1016/j.jacl.2022.10.004] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Revised: 10/11/2022] [Accepted: 10/12/2022] [Indexed: 11/05/2022]
Abstract
The role of lipoprotein(a) (Lp[a]) as a significant and possibly causal cardiovascular disease (CVD) risk factor has been well established. Many studies, mostly experimental, have supported inflammation as a mediator of Lp(a)-induced increase in CVD risk. Lp(a), mainly through oxidized phospholipids bound to its apolipoprotein(a) part, leads to monocyte activation and endothelial dysfunction. The relationship between Lp(a) and inflammation is bidirectional as Lp(a) levels, besides being associated with inflammatory properties, are regulated by inflammatory stimuli or anti-inflammatory treatment. Reduction of Lp(a) concentration, especially by potent siRNA agents, contributes to partial reversion of the Lp(a) related inflammatory profile. This review aims to present the current pathophysiological and clinical evidence of the relationship between Lp(a) and inflammation.
Collapse
Affiliation(s)
- Spyridon Simantiris
- 1st Cardiology Department, Hippokration Hospital, National and Kapodistrian University of Athens, Vas. Sofias Avenue 114, Athens 11527, Greece (Drs Simantiris, Antonopoulos, Papastamos, Benetos, Tsioufis, and Tousoulis)
| | - Alexios S Antonopoulos
- 1st Cardiology Department, Hippokration Hospital, National and Kapodistrian University of Athens, Vas. Sofias Avenue 114, Athens 11527, Greece (Drs Simantiris, Antonopoulos, Papastamos, Benetos, Tsioufis, and Tousoulis)
| | - Charalampos Papastamos
- 1st Cardiology Department, Hippokration Hospital, National and Kapodistrian University of Athens, Vas. Sofias Avenue 114, Athens 11527, Greece (Drs Simantiris, Antonopoulos, Papastamos, Benetos, Tsioufis, and Tousoulis)
| | - Georgios Benetos
- 1st Cardiology Department, Hippokration Hospital, National and Kapodistrian University of Athens, Vas. Sofias Avenue 114, Athens 11527, Greece (Drs Simantiris, Antonopoulos, Papastamos, Benetos, Tsioufis, and Tousoulis)
| | - Nikolaos Koumallos
- Department of Cardiothoracic Surgery, Hippokration Hospital, Athens, Greece (Dr Koumallos)
| | - Konstantinos Tsioufis
- 1st Cardiology Department, Hippokration Hospital, National and Kapodistrian University of Athens, Vas. Sofias Avenue 114, Athens 11527, Greece (Drs Simantiris, Antonopoulos, Papastamos, Benetos, Tsioufis, and Tousoulis)
| | - Dimitris Tousoulis
- 1st Cardiology Department, Hippokration Hospital, National and Kapodistrian University of Athens, Vas. Sofias Avenue 114, Athens 11527, Greece (Drs Simantiris, Antonopoulos, Papastamos, Benetos, Tsioufis, and Tousoulis).
| |
Collapse
|
19
|
Antidyslipidemic, Antioxidant, and Anti-inflammatory Effects of Jelly Drink Containing Polyphenol-Rich Roselle Calyces Extract and Passion Fruit Juice with Pulp in Adults with Dyslipidemia: A Randomized, Double-Blind, Placebo-Controlled Trial. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:4631983. [PMID: 36187334 PMCID: PMC9519340 DOI: 10.1155/2022/4631983] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Accepted: 09/01/2022] [Indexed: 11/21/2022]
Abstract
Oxidative stress and inflammation play key roles in the pathophysiology in the pathophysiology of dyslipidemia, which are positive risks that increase atherosclerosis leading to important healthcare problems. Therefore, we aimed to study the antioxidant, anti-inflammatory, and lipid-lowering effects of jelly drink containing polyphenol-rich roselle calyces extract and passion fruit juice with pulp concentrate (RP jelly drink) in comparison to a placebo jelly drink for 8 weeks. Forty-three adults with dyslipidemia were randomly assigned into two groups: the RP jelly drink group and the placebo group. Glucose, total cholesterol (TC) triglyceride (TG), low-density lipoprotein-cholesterol (LDL-C), high-density lipoprotein-cholesterol (HDL-C), oxidative stress biomarkers, inflammatory parameters, and monocyte chemotactic protein-1 (MCP-1) were measured with fasting blood samples at baseline, 4 weeks and 8 weeks of intervention. Results showed a significant decrease in LDL-C and TG, respectively, after 8 weeks of RP jelly drink consumption (LDL-C: 107.63 ± 22.98 mg/dL; TG: 109.79 ± 38.83 mg/dL) compared to baseline measurements (LDL-C: 128.43 ± 32.74 mg/dL; TG: 132.33 ± 75.11 mg/dL). These may be possible due to reduced inflammation and improvements in oxidative stress, as demonstrated by the reduction of tumor necrosis factor- (TNF-) α and malondialdehyde (MDA), and the enhancement of glutathione (GSH) after consuming the RP jelly drink for 8 weeks. However, no significant differences of treatment on glucose, total cholesterol, MCP-1, interleukin-6, and interleukin-10 were observed. In conclusion, daily consumption of RP jelly drink for 8 weeks resulted in significant improvement in lipid profiles in subjects with dyslipidemia. However, more research is needed to assess its nutritional and functional potential.
Collapse
|
20
|
O’Hagan R, Berg AR, Hong CG, Parel PM, Mehta NN, Teague HL. Systemic consequences of abnormal cholesterol handling: Interdependent pathways of inflammation and dyslipidemia. Front Immunol 2022; 13:972140. [PMID: 36091062 PMCID: PMC9459038 DOI: 10.3389/fimmu.2022.972140] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Accepted: 08/05/2022] [Indexed: 11/13/2022] Open
Abstract
Metabolic conditions such as obesity and associated comorbidities are increasing in prevalence worldwide. In chronically inflamed pathologies, metabolic conditions are linked to early onset cardiovascular disease, which remains the leading cause of death despite decades of research. In recent years, studies focused on the interdependent pathways connecting metabolism and the immune response have highlighted that dysregulated cholesterol trafficking instigates an overactive, systemic inflammatory response, thereby perpetuating early development of cardiovascular disease. In this review, we will discuss the overlapping pathways connecting cholesterol trafficking with innate immunity and present evidence that cholesterol accumulation in the bone marrow may drive systemic inflammation in chronically inflamed pathologies. Lastly, we will review the current therapeutic strategies that target both inflammation and cholesterol transport, and how biologic therapy restores lipoprotein function and mitigates the immune response.
Collapse
|
21
|
Zhang S, Xie S, Gao Y, Wang Y. Triptolide alleviates ox-LDL-induced endothelial inflammation via attenuating the oxidative stress-mediated NF-κB pathway. Curr Ther Res Clin Exp 2022; 97:100683. [PMID: 35989981 PMCID: PMC9385561 DOI: 10.1016/j.curtheres.2022.100683] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2022] [Accepted: 07/13/2022] [Indexed: 11/20/2022] Open
Abstract
Background Endothelial inflammation triggered by oxidized LDL (ox-LDL) is a crucial mechanism involved in atherosclerosis. Triptolide (TP), a primary active ingredient of the traditional Chinese medicine Tripterygium wilfordii Hook F, possesses antioxidant and anti-inflammatory properties in vivo. However, limited information is available regarding these effects on endothelial inflammation occurring in atherosclerosis. Objectives This study investigated the effects and possible mechanisms of action of TP on ox–LDL-induced inflammatory responses in human umbilical vein endothelial cells. Methods Human umbilical vein endothelial cells were preincubated with TP at the indicated concentrations for 1 hour and then incubated with ox-LDL (50 µg/mL) for the indicated times. Results Preincubation of cultured human umbilical vein endothelial cells with TP inhibited ox–LDL-induced cytokine and chemokine production, adhesion molecule expression, and monocyte adhesion in a concentration-dependent manner. The concentrations of 8-isoprostane, malondialdehyde, and superoxide increased after human umbilical vein endothelial cells were exposed to ox-LDL, which were associated with decreased activities of total superoxide dismutase and its isoenzyme (ie, CuZn- superoxide dismutase). Preincubation with TP reversed ox–LDL-induced effects in all events. Moreover, preincubation with TP also attenuated ox–LDL-induced nuclear factor-kappa B transcriptional activation in a concentration-dependent manner, via the suppression of inhibitor of kappa Balpha (IκBα) phosphorylation and subsequent nuclear factor-kappa B DNA binding. Conclusions These data indicate that TP inhibits ox–LDL-induced endothelial inflammation, possibly via suppression of the oxidative stress-dependent activation of the nuclear factor-kappa B signaling pathway.
Collapse
Affiliation(s)
| | | | | | - Youping Wang
- Address correspondence to: Youping Wang, MD, PhD, Central Laboratory and Division of Cardiology, First Affiliated Hospital, Henan University of Traditional Chinese Medicine, Zhengzhou, 450000, China.
| |
Collapse
|
22
|
Olechno E, Puścion-Jakubik A, Zujko ME. Chokeberry (A. melanocarpa (Michx.) Elliott)—A Natural Product for Metabolic Disorders? Nutrients 2022; 14:nu14132688. [PMID: 35807867 PMCID: PMC9268775 DOI: 10.3390/nu14132688] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2022] [Revised: 06/25/2022] [Accepted: 06/25/2022] [Indexed: 11/16/2022] Open
Abstract
Abnormal metabolism of substances in the body can result in metabolic disorders which include obesity, cardiovascular diseases, diabetes, hypertension, chronic kidney disease, liver disease, or cancer. Foods rich in antioxidants can help to prevent and treat various types of disorders. Chokeberry fruits are rich in polyphenols, especially cyanidins, and therefore, can show a beneficial health effect. The aim of this study was to summarize and systematize reports about the effects of chokeberry on various metabolic parameters. Studies from 2000 to 2021, published in the PubMed and Google Scholar databases, were reviewed. The review of studies shows that chokeberry may have a positive effect in dyslipidemia and hypertension and may increase the body’s antioxidant defense mechanisms. The anti-inflammatory effect, in turn, may translate into a reduction in the risk of metabolic disorders over a longer period of use. Changes in glucose levels were reported by studies in which the intervention lasted more than 10 weeks in patients with carbohydrate metabolism disorders. The effects of protecting the liver, inhibiting platelet aggregation, lowering uric acid levels, and having a protective effect on the kidneys require additional confirmation in human clinical trials. Consumption of chokeberry fruit did not impact on anthropometric measurements; however, it seems that chokeberry fruit can be recommended in many metabolic disorders due to the richness of bioactive ingredients.
Collapse
Affiliation(s)
- Ewa Olechno
- Department of Food Biotechnology, Faculty of Health Science, Medical University of Białystok, Szpitalna 37 Street, 15-295 Białystok, Poland; (E.O.); (M.E.Z.)
| | - Anna Puścion-Jakubik
- Department of Bromatology, Faculty of Pharmacy with the Division of Laboratory Medicine, Medical University of Białystok, Mickiewicza 2D Street, 15-222 Białystok, Poland
- Correspondence: ; Tel.: +48-85-748-54-69
| | - Małgorzata Elżbieta Zujko
- Department of Food Biotechnology, Faculty of Health Science, Medical University of Białystok, Szpitalna 37 Street, 15-295 Białystok, Poland; (E.O.); (M.E.Z.)
| |
Collapse
|
23
|
Hu J, Lei H, Liu L, Xu D. Lipoprotein(a), a Lethal Player in Calcific Aortic Valve Disease. Front Cell Dev Biol 2022; 10:812368. [PMID: 35155427 PMCID: PMC8830536 DOI: 10.3389/fcell.2022.812368] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Accepted: 01/07/2022] [Indexed: 12/11/2022] Open
Abstract
Calcified aortic valve disease (CAVD) is the most common valvular cardiovascular disease with increasing incidence and mortality. The primary treatment for CAVD is surgical or transcatheter aortic valve replacement and there remains a lack of effective drug treatment. Recently, lipoprotein (a) (Lp(a)) has been considered to play a crucial role in CAVD pathophysiology. Multiple studies have shown that Lp(a) represents an independent risk factor for CAVD. Moreover, Lp(a) mediates the occurrence and development of CAVD by affecting aortic valve endothelial dysfunction, indirectly promoting foam cell formation through oxidized phospholipids (OxPL), inflammation, oxidative stress, and directly promotes valve calcification. However, there is a lack of clinical trials with Lp(a) reduction as a primary endpoint. This review aims to explore the relationship and mechanism between Lp(a) and CAVD, and focuses on the current drugs that can be used as potential therapeutic targets for CAVD.
Collapse
Affiliation(s)
- Jiahui Hu
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha, China
- Research Institute of Blood Lipid and Atherosclerosis, Central South University, Changsha, China
- Modern Cardiovascular Disease Clinical Technology Research Center of Hunan Province, Changsha, China
- Cardiovascular Disease Research Center of Hunan Province, Changsha, China
| | - Hao Lei
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha, China
- Research Institute of Blood Lipid and Atherosclerosis, Central South University, Changsha, China
- Modern Cardiovascular Disease Clinical Technology Research Center of Hunan Province, Changsha, China
- Cardiovascular Disease Research Center of Hunan Province, Changsha, China
| | - Leiling Liu
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha, China
- Research Institute of Blood Lipid and Atherosclerosis, Central South University, Changsha, China
- Modern Cardiovascular Disease Clinical Technology Research Center of Hunan Province, Changsha, China
- Cardiovascular Disease Research Center of Hunan Province, Changsha, China
| | - Danyan Xu
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha, China
- Research Institute of Blood Lipid and Atherosclerosis, Central South University, Changsha, China
- Modern Cardiovascular Disease Clinical Technology Research Center of Hunan Province, Changsha, China
- Cardiovascular Disease Research Center of Hunan Province, Changsha, China
- *Correspondence: Danyan Xu,
| |
Collapse
|
24
|
Ugovšek S, Šebeštjen M. Lipoprotein(a)—The Crossroads of Atherosclerosis, Atherothrombosis and Inflammation. Biomolecules 2021; 12:biom12010026. [PMID: 35053174 PMCID: PMC8773759 DOI: 10.3390/biom12010026] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Revised: 12/22/2021] [Accepted: 12/23/2021] [Indexed: 12/14/2022] Open
Abstract
Increased lipoprotein(a) (Lp(a)) levels are an independent predictor of coronary artery disease (CAD), degenerative aortic stenosis (DAS), and heart failure independent of CAD and DAS. Lp(a) levels are genetically determinated in an autosomal dominant mode, with great intra- and inter-ethnic diversity. Most variations in Lp(a) levels arise from genetic variations of the gene that encodes the apolipoprotein(a) component of Lp(a), the LPA gene. LPA is located on the long arm of chromosome 6, within region 6q2.6–2.7. Lp(a) levels increase cardiovascular risk through several unrelated mechanisms. Lp(a) quantitatively carries all of the atherogenic risk of low-density lipoprotein cholesterol, although it is even more prone to oxidation and penetration through endothelia to promote the production of foam cells. The thrombogenic properties of Lp(a) result from the homology between apolipoprotein(a) and plasminogen, which compete for the same binding sites on endothelial cells to inhibit fibrinolysis and promote intravascular thrombosis. LPA has up to 70% homology with the human plasminogen gene. Oxidized phospholipids promote differentiation of pro-inflammatory macrophages that secrete pro-inflammatory cytokines (e. g., interleukin (IL)-1β, IL-6, IL-8, tumor necrosis factor-α). The aim of this review is to define which of these mechanisms of Lp(a) is predominant in different groups of patients.
Collapse
Affiliation(s)
- Sabina Ugovšek
- Faculty of Medicine, University of Ljubljana, 1000 Ljubljana, Slovenia;
| | - Miran Šebeštjen
- Faculty of Medicine, University of Ljubljana, 1000 Ljubljana, Slovenia;
- Department of Cardiology, University Medical Centre Ljubljana, 1000 Ljubljana, Slovenia
- Department of Vascular Diseases, University Medical Centre Ljubljana, 1000 Ljubljana, Slovenia
- Correspondence:
| |
Collapse
|
25
|
Abstract
PURPOSE OF REVIEW Lipid-mediated atherogenesis is hallmarked by a chronic inflammatory state. Low-density lipoprotein cholesterol (LDL-C), triglyceride rich lipoproteins (TRLs), and lipoprotein(a) [Lp(a)] are causally related to atherosclerosis. Within the paradigm of endothelial activation and subendothelial lipid deposition, these lipoproteins induce numerous pro-inflammatory pathways. In this review, we will outline the effects of lipoproteins on systemic inflammatory pathways in atherosclerosis. RECENT FINDINGS Apolipoprotein B-containing lipoproteins exert a variety of pro-inflammatory effects, ranging from the local artery to systemic immune cell activation. LDL-C, TRLs, and Lp(a) induce endothelial dysfunction with concomitant activation of circulating monocytes through enhanced lipid accumulation. The process of trained immunity of the innate immune system, predominantly induced by LDL-C particles, hallmarks the propagation of the low-grade inflammatory response. In concert, bone marrow activation induces myeloid skewing, further contributing to immune cell mobilization and plaque progression. SUMMARY Lipoproteins and inflammation are intertwined in atherogenesis. Elucidating the inflammatory pathways will provide new opportunities for therapeutic agents.
Collapse
Affiliation(s)
- Jordan M. Kraaijenhof
- Department of Vascular Medicine, Amsterdam UMC, Location AMC, University of Amsterdam
| | - G. Kees Hovingh
- Department of Vascular Medicine, Amsterdam UMC, Location AMC, University of Amsterdam
| | - Erik S.G. Stroes
- Department of Vascular Medicine, Amsterdam UMC, Location AMC, University of Amsterdam
| | - Jeffrey Kroon
- Amsterdam UMC, University of Amsterdam, Department of Experimental Vascular Medicine, Amsterdam Cardiovascular Sciences, Amsterdam, The Netherlands
| |
Collapse
|
26
|
Hou Y, Guo W, Fan T, Li B, Ge W, Gao R, Wang J. Advanced Research of Abdominal Aortic Aneurysms on Metabolism. Front Cardiovasc Med 2021; 8:630269. [PMID: 33614752 PMCID: PMC7892590 DOI: 10.3389/fcvm.2021.630269] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Accepted: 01/05/2021] [Indexed: 01/16/2023] Open
Abstract
Abdominal aortic aneurysm (AAA) is a cardiovascular disease with a high risk of death, seriously threatening the life and health of people. The specific pathogenesis of AAA is still not fully understood. In recent years, researchers have found that amino acid, lipid, and carbohydrate metabolism disorders play important roles in the occurrence and development of AAA. This review is aimed to summarize the latest research progress of the relationship between AAA progression and body metabolism. The body metabolism is closely related to the occurrence and development of AAA. It is necessary to further investigate the pathogenesis of AAA from the perspective of metabolism to provide theoretical basis for AAA diagnosis and drug development.
Collapse
Affiliation(s)
- Yangfeng Hou
- State Key Laboratory of Medical Molecular Biology, Institute of Basic Medicine, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, Beijing, China
| | - Wenjun Guo
- State Key Laboratory of Medical Molecular Biology, Institute of Basic Medicine, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, Beijing, China
| | - Tianfei Fan
- State Key Laboratory of Medical Molecular Biology, Institute of Basic Medicine, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, Beijing, China
| | - Bolun Li
- State Key Laboratory of Medical Molecular Biology, Institute of Basic Medicine, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, Beijing, China
| | - Weipeng Ge
- State Key Laboratory of Medical Molecular Biology, Institute of Basic Medicine, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, Beijing, China
| | - Ran Gao
- State Key Laboratory of Medical Molecular Biology, Institute of Basic Medicine, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, Beijing, China
| | - Jing Wang
- State Key Laboratory of Medical Molecular Biology, Institute of Basic Medicine, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, Beijing, China
| |
Collapse
|
27
|
Ke G, Hans C, Agarwal G, Orion K, Go M, Hao W. Mathematical model of atherosclerotic aneurysm. MATHEMATICAL BIOSCIENCES AND ENGINEERING : MBE 2021; 18:1465-1484. [PMID: 33757194 DOI: 10.3934/mbe.2021076] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Atherosclerosis is a major cause of abdominal aortic aneurysm (AAA) and up to 80% of AAA patients have atherosclerosis. Therefore it is critical to understand the relationship and interactions between atherosclerosis and AAA to treat atherosclerotic aneurysm patients more effectively. In this paper, we develop a mathematical model to mimic the progression of atherosclerotic aneurysms by including both the multi-layer structured arterial wall and the pathophysiology of atherosclerotic aneurysms. The model is given by a system of partial differential equations with free boundaries. Our results reveal a 2D biomarker, the cholesterol ratio and DDR1 level, assessing the risk of atherosclerotic aneurysms. The efficacy of different treatment plans is also explored via our model and suggests that the dosage of anti-cholesterol drugs is significant to slow down the progression of atherosclerotic aneurysms while the additional anti-DDR1 injection can further reduce the risk.
Collapse
Affiliation(s)
- Guoyi Ke
- Department of Mathematics and Physical Sciences, Louisiana State University at Alexandria, Alexandria, LA 71302, USA
| | - Chetan Hans
- School of Medicine, University of Missouri, Columbia, MO 65212, USA
| | - Gunjan Agarwal
- Department of Mechanical Aerospace Engineering, Ohio State University, Columbus, OH 43210-1142, USA
| | - Kristine Orion
- Ohio State Uniersity Wexner Medical Center, Columbus, OH 43210-1142, USA
| | - Michael Go
- Ohio State Uniersity Wexner Medical Center, Columbus, OH 43210-1142, USA
| | - Wenrui Hao
- Department of Mathematics, Pennsylvania State University, PA 16802, USA
| |
Collapse
|
28
|
Liu T, Yoon WS, Lee SR. Recent Updates of Lipoprotein(a) and Cardiovascular Disease. Chonnam Med J 2021; 57:36-43. [PMID: 33537217 PMCID: PMC7840349 DOI: 10.4068/cmj.2021.57.1.36] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Revised: 12/14/2020] [Accepted: 12/16/2020] [Indexed: 12/22/2022] Open
Abstract
In recent years, epidemiological studies, genome-wide association studies, and Mendelian randomization studies have shown a strong association between increased levels of lipoproteins and increased risks of coronary heart disease and cardiovascular disease (CVD). Although lipoprotein(a) [Lp(a)] was an independent risk factor for ASCVD, the latest international clinical guidelines do not recommend direct reduction of plasma Lp(a) concentrations. The main reason was that there is no effective clinical medicine that directly lowers plasma Lp(a) concentrations. However, recent clinical trials have shown that proprotein convertase subtilisin/kexin-type 9 inhibitors (PCSK9) and second-generation antisense oligonucleotides can effectively reduce plasma Lp(a) levels. This review will present the structure, pathogenicity, prognostic evidences, and recent advances in therapeutic drugs for Lp(a).
Collapse
Affiliation(s)
- Taili Liu
- Division of Cardiology, Department of Internal Medicine, Chonbuk National University Hospital, Jeonju, Korea
| | - Won-Sik Yoon
- Division of Cardiology, Department of Internal Medicine, Chonbuk National University Hospital, Jeonju, Korea
| | - Sang-Rok Lee
- Division of Cardiology, Department of Internal Medicine, Chonbuk National University Hospital, Jeonju, Korea
| |
Collapse
|
29
|
Bourgeois R, Girard A, Perrot N, Guertin J, Mitchell PL, Couture C, Gotti C, Bourassa S, Poggio P, Mass E, Capoulade R, Scipione CA, Després AA, Couture P, Droit A, Pibarot P, Boffa MB, Thériault S, Koschinsky ML, Mathieu P, Arsenault BJ. A Comparative Analysis of the Lipoprotein(a) and Low-Density Lipoprotein Proteomic Profiles Combining Mass Spectrometry and Mendelian Randomization. CJC Open 2020; 3:450-459. [PMID: 34027348 PMCID: PMC8129481 DOI: 10.1016/j.cjco.2020.11.019] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Accepted: 11/28/2020] [Indexed: 12/30/2022] Open
Abstract
Background Lipoprotein(a) (Lp[a]), which consists of a low-density lipoprotein (LDL) bound to apolipoprotein(a), is one of the strongest genetic risk factors for atherosclerotic cardiovascular diseases. Few studies have performed hypothesis-free direct comparisons of the Lp(a) and the LDL proteomes. Our objectives were to compare the Lp(a) and the LDL proteomic profiles and to evaluate the effect of lifelong exposure to elevated Lp(a) or LDL cholesterol levels on the plasma proteomic profile. Methods We performed a label-free analysis of the Lp(a) and LDL proteomic profiles of healthy volunteers in a discovery (n = 6) and a replication (n = 9) phase. We performed inverse variance weighted Mendelian randomization to document the effect of lifelong exposure to elevated Lp(a) or LDL cholesterol levels on the plasma proteomic profile of participants of the INTERVAL study. Results We identified 15 proteins that were more abundant on Lp(a) compared with LDL (serping1, pi16, itih1, itih2, itih3, pon1, podxl, cd44, cp, ptprg, vtn, pcsk9, igfals, vcam1, and ttr). We found no proteins that were more abundant on LDL compared with Lp(a). After correction for multiple testing, lifelong exposure to elevated LDL cholesterol levels was associated with the variation of 18 plasma proteins whereas Lp(a) did not appear to influence the plasma proteome. Conclusions Results of this study highlight marked differences in the proteome of Lp(a) and LDL as well as in the effect of lifelong exposure to elevated LDL cholesterol or Lp(a) on the plasma proteomic profile.
Collapse
Affiliation(s)
- Raphaëlle Bourgeois
- Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec, Quebec, Canada.,Department of Medicine, Faculty of Medicine, Université Laval, Quebec, Canada
| | - Arnaud Girard
- Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec, Quebec, Canada.,Department of Medicine, Faculty of Medicine, Université Laval, Quebec, Canada
| | - Nicolas Perrot
- Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec, Quebec, Canada.,Department of Medicine, Faculty of Medicine, Université Laval, Quebec, Canada
| | - Jakie Guertin
- Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec, Quebec, Canada.,Department of Medicine, Faculty of Medicine, Université Laval, Quebec, Canada
| | - Patricia L Mitchell
- Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec, Quebec, Canada
| | - Christian Couture
- Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec, Quebec, Canada
| | - Clarisse Gotti
- Proteomics platform of the CHU de Québec, Quebec, Canada
| | | | | | - Elvira Mass
- University of Bonn, Developmental Biology of the Immune System, Life and Medical Sciences Institute (LIMES), Bonn, Germany
| | - Romain Capoulade
- Université de Nantes, CHU Nantes, CNRS, INSERM, l'institut du thorax, Nantes, France
| | - Corey A Scipione
- Robarts Research Institute, Schulich School of Medicine and Dentistry, The University of Western Ontario, London, Ontario, Canada
| | - Audrey-Anne Després
- Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec, Quebec, Canada.,Department of Medicine, Faculty of Medicine, Université Laval, Quebec, Canada
| | - Patrick Couture
- Department of Medicine, Faculty of Medicine, Université Laval, Quebec, Canada.,Centre de recherche du CHU de Québec, Quebec, Canada
| | - Arnaud Droit
- Proteomics platform of the CHU de Québec, Quebec, Canada.,Centre de recherche du CHU de Québec, Quebec, Canada
| | - Philippe Pibarot
- Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec, Quebec, Canada.,Department of Medicine, Faculty of Medicine, Université Laval, Quebec, Canada
| | - Michael B Boffa
- Department of Biochemistry, Schulich School of Medicine and Dentistry, The University of Western Ontario, London, Ontario, Canada
| | - Sébastien Thériault
- Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec, Quebec, Canada.,Department of Molecular Biology, Medical Biochemistry and Pathology, Faculty of Medicine, Université Laval, Quebec, Canada
| | - Marlys L Koschinsky
- Robarts Research Institute, Schulich School of Medicine and Dentistry, The University of Western Ontario, London, Ontario, Canada
| | - Patrick Mathieu
- Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec, Quebec, Canada.,Department of Surgery, Faculty of Medicine, Université Laval, Quebec, Canada
| | - Benoit J Arsenault
- Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec, Quebec, Canada.,Department of Medicine, Faculty of Medicine, Université Laval, Quebec, Canada
| |
Collapse
|
30
|
Xuan L, Wang T, Dai H, Wang B, Xiang J, Wang S, Lin H, Li M, Zhao Z, Lu J, Chen Y, Xu Y, Wang W, Xu M, Bi Y, Ning G. Serum lipoprotein (a) associates with a higher risk of reduced renal function: a prospective investigation. J Lipid Res 2020; 61:1320-1327. [PMID: 32703886 PMCID: PMC7529054 DOI: 10.1194/jlr.ra120000771] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Lipoprotein (a) [Lp(a)] is a well-known risk factor for cardiovascular disease, but analysis on Lp(a) and renal dysfunction is scarce. We aimed to investigate prospectively the association of serum Lp(a) with the risk of reduced renal function, and further investigated whether diabetic or hypertensive status modified such association. Six thousand two hundred and fifty-seven Chinese adults aged ≤40 years and free of reduced renal function at baseline were included in the study. Reduced renal function was defined as estimated glomerular filtration rate <60 ml/min/1.73 m2 During a mean follow-up of 4.4 years, 158 participants developed reduced renal function. Each one-unit increase in log10-Lp(a) (milligrams per deciliter) was associated with a 1.99-fold (95% CI 1.15-3.43) increased risk of incident reduced renal function; the multivariable-adjusted odds ratio (OR) for the highest tertile of Lp(a) was 1.61 (95% CI 1.03-2.52) compared with the lowest tertile (P for trend = 0.03). The stratified analysis showed the association of serum Lp(a) and incident reduced renal function was more prominent in participants with prevalent diabetes [OR 4.04, 95% CI (1.42-11.54)] or hypertension [OR 2.18, 95% CI (1.22-3.89)]. A stronger association was observed in the group with diabetes and high Lp(a) (>25 mg/dl), indicating a combined effect of diabetes and high Lp(a) on the reduced renal function risk. An elevated Lp(a) level was independently associated with risk of incident reduced renal function, especially in diabetic or hypertensive patients.
Collapse
Affiliation(s)
- Liping Xuan
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China, and Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Tiange Wang
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China, and Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Huajie Dai
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China, and Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Bin Wang
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China, and Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jiali Xiang
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China, and Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Shuangyuan Wang
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China, and Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hong Lin
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China, and Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Mian Li
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China, and Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhiyun Zhao
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China, and Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jieli Lu
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China, and Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yuhong Chen
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China, and Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yu Xu
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China, and Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Weiqing Wang
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China, and Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Min Xu
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China, and Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yufang Bi
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China, and Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Guang Ning
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China, and Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
31
|
Zou J, Wang G, Li H, Yu X, Tang C. IgM natural antibody T15/E06 in atherosclerosis. Clin Chim Acta 2020; 504:15-22. [DOI: 10.1016/j.cca.2020.01.024] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2019] [Revised: 01/23/2020] [Accepted: 01/23/2020] [Indexed: 11/28/2022]
|
32
|
Jan MI, Ali T, Ishtiaq A, Mushtaq I, Murtaza I. Prospective Advances in Non-coding RNAs Investigation. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1229:385-426. [PMID: 32285426 DOI: 10.1007/978-981-15-1671-9_24] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 12/09/2022]
Abstract
Non-coding RNAs (ncRNAs) play significant roles in numerous physiological cellular processes and molecular alterations during pathological conditions including heart diseases, cancer, immunological disorders and neurological diseases. This chapter is focusing on the basis of ncRNA relation with their functions and prospective advances in non-coding RNAs particularly miRNAs investigation in the cardiovascular disease management.The field of ncRNAs therapeutics is a very fascinating and challenging too. Scientists have opportunity to develop more advanced therapeutics as well as diagnostic approaches for cardiovascular conditions. Advanced studies are critically needed to deepen the understanding of the molecular biology, mechanism and modulation of ncRNAs and chemical formulations for managing CVDs.
Collapse
Affiliation(s)
- Muhammad Ishtiaq Jan
- Department of Biochemistry, Signal Transduction Laboratory, Faculty of Biological Sciences, Quaid-i-Azam University, Islamabad, Pakistan
| | - Tahir Ali
- Department of Biochemistry, Signal Transduction Laboratory, Faculty of Biological Sciences, Quaid-i-Azam University, Islamabad, Pakistan
| | - Ayesha Ishtiaq
- Department of Biochemistry, Signal Transduction Laboratory, Faculty of Biological Sciences, Quaid-i-Azam University, Islamabad, Pakistan
| | - Iram Mushtaq
- Department of Biochemistry, Signal Transduction Laboratory, Faculty of Biological Sciences, Quaid-i-Azam University, Islamabad, Pakistan
| | - Iram Murtaza
- Department of Biochemistry, Signal Transduction Laboratory, Faculty of Biological Sciences, Quaid-i-Azam University, Islamabad, Pakistan.
| |
Collapse
|
33
|
Sabouret P, Angoulvant D, Ray KK. Lipoprotein(a), the rediscovered risk factor, or how to get "back to the future". Arch Cardiovasc Dis 2020; 113:147-151. [PMID: 32205042 DOI: 10.1016/j.acvd.2020.03.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/05/2019] [Revised: 03/04/2020] [Accepted: 03/05/2020] [Indexed: 11/25/2022]
Affiliation(s)
- Pierre Sabouret
- Heart Institute, Pitié-Salpétrière Hospital, ACTION-Group, Pierre and Marie Curie University, 47-83 boulevard de L'Hôpital, 75013 Paris, France.
| | - Denis Angoulvant
- Department of Cardiology, CHRU de Tours and EA4245 T2i, Loire Valley Cardiovascular Collaboration, Tours University, 37000 Tours, France
| | - Kausik K Ray
- Department of Public Health, Imperial College, W6 8RP London, UK
| |
Collapse
|
34
|
Jawi MM, Frohlich J, Chan SY. Lipoprotein(a) the Insurgent: A New Insight into the Structure, Function, Metabolism, Pathogenicity, and Medications Affecting Lipoprotein(a) Molecule. J Lipids 2020; 2020:3491764. [PMID: 32099678 PMCID: PMC7016456 DOI: 10.1155/2020/3491764] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2019] [Accepted: 08/17/2019] [Indexed: 12/15/2022] Open
Abstract
Lipoprotein(a) [Lp(a)], aka "Lp little a", was discovered in the 1960s in the lab of the Norwegian physician Kåre Berg. Since then, we have greatly improved our knowledge of lipids and cardiovascular disease (CVD). Lp(a) is an enigmatic class of lipoprotein that is exclusively formed in the liver and comprises two main components, a single copy of apolipoprotein (apo) B-100 (apo-B100) tethered to a single copy of a protein denoted as apolipoprotein(a) apo(a). Plasma levels of Lp(a) increase soon after birth to a steady concentration within a few months of life. In adults, Lp(a) levels range widely from <2 to 2500 mg/L. Evidence that elevated Lp(a) levels >300 mg/L contribute to CVD is significant. The improvement of isoform-independent assays, together with the insight from epidemiologic studies, meta-analyses, genome-wide association studies, and Mendelian randomization studies, has established Lp(a) as the single most common independent genetically inherited causal risk factor for CVD. This breakthrough elevated Lp(a) from a biomarker of atherosclerotic risk to a target of therapy. With the emergence of promising second-generation antisense therapy, we hope that we can answer the question of whether Lp(a) is ready for prime-time clinic use. In this review, we present an update on the metabolism, pathophysiology, and current/future medical interventions for high levels of Lp(a).
Collapse
Affiliation(s)
- Motasim M. Jawi
- Healthy Heart Program, St. Paul's Hospital, Vancouver V6Z 1Y6, Canada
- Division of Experimental Medicine, Department of Medicine, Faculty of Medicine, University of British Columbia, Vancouver V5Z 1M9, Canada
- Department of Clinical PhysiologyCorrection: Department of Physiology, University of Jeddah, P.O. Box: 24, Jeddah 21959, Saudi Arabia
| | - Jiri Frohlich
- Healthy Heart Program, St. Paul's Hospital, Vancouver V6Z 1Y6, Canada
- Department of Pathology and Laboratory Medicine, Faculty of Medicine, University of British Columbia, Vancouver, British Columbia V6T 2B5, Canada
| | - Sammy Y. Chan
- Healthy Heart Program, St. Paul's Hospital, Vancouver V6Z 1Y6, Canada
- Department of Medicine, Division of Cardiology, University of British Columbia, Vancouver V5Z 1M9, Canada
| |
Collapse
|
35
|
Cai L, Zhang X, Hou M, Gao F. Natural flavone tricetin suppresses oxidized LDL-induced endothelial inflammation mediated by Egr-1. Int Immunopharmacol 2020; 80:106224. [PMID: 31991371 DOI: 10.1016/j.intimp.2020.106224] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2019] [Revised: 01/02/2020] [Accepted: 01/16/2020] [Indexed: 02/08/2023]
Abstract
Atherosclerosis is the primary cause of many cardiovascular diseases. Endothelial dysfunction is recognized as a crucial early event in atherosclerotic lesion formation. Tricetin is a natural flavonoid derivative that has demonstrated a wide range of therapeutic properties. This study investigates the protective effect of tricetin in cultured endothelial cells. The results of our study show that tricetin suppressed oxidized low-density lipoprotein (ox-LDL)-induced expression of pro-inflammatory monocyte chemotactic protein-1 (MCP-1) and interleukin-1β (IL-1β), as well as the generation of reactive oxygen species (ROS). Furthermore, our findings indicate that tricetin suppressed ox-LDL-induced expression of intercellular adhesion molecule-1 (ICAM-1) and vascular cell adhesion molecule-1 (VCAM-1). At the cellular level, the presence of tricetin inhibited ox-LDL-induced monocyte adhesion to endothelial cells. Mechanistically, we showed that tricetin suppressed the induction of the endothelial receptor for ox-LDL, lectin-like ox-LDL receptor-1 (LOX-1), and the transcriptional factor early growth response 1 (Egr-1) as well as extracellular signal-regulated protein kinase 1 and 2 (ERK1/2) activation. These data demonstrate that tricetin is a natural protective agent in vascular endothelial cells, indicating that tricetin could have a potentially beneficial effect in the modulation of atherosclerosis.
Collapse
Affiliation(s)
- Luming Cai
- Department of Cardiovascular Medicine, Jingjiang People's Hospital, Jingjiang, Jiangsu 214500, PR China
| | - Xuesong Zhang
- Department of Central Laboratory, Jingjiang People's Hospital, Jingjiang, Jiangsu 214500, PR China
| | - Meiling Hou
- Department of Cardiovascular Medicine, Jingjiang People's Hospital, Jingjiang, Jiangsu 214500, PR China
| | - Feng Gao
- Hospital Office, Jingjiang People's Hospital, Jingjiang, Jiangsu 214500, PR China.
| |
Collapse
|
36
|
Umar S, Ruffenach G, Moazeni S, Vaillancourt M, Hong J, Cunningham C, Cao N, Navab S, Sarji S, Li M, Lee L, Fishbein G, Ardehali A, Navab M, Reddy ST, Eghbali M. Involvement of Low-Density Lipoprotein Receptor in the Pathogenesis of Pulmonary Hypertension. J Am Heart Assoc 2020; 9:e012063. [PMID: 31914876 PMCID: PMC7033825 DOI: 10.1161/jaha.119.012063] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Background Recently, we and others have reported a causal role for oxidized lipids in the pathogenesis of pulmonary hypertension (PH). However, the role of low‐density lipoprotein receptor (LDL‐R) in PH is not known. Methods and Results We examined the role of LDL‐R in the development of PH and determined the efficacy of high‐density lipoprotein mimetic peptide 4F in mitigating PH. Explanted human lungs and plasma from patients with PH and control subjects were analyzed for gene expression, histological characteristics, and lipoprotein oxidation. Male LDL‐R null (LDL‐R knockout) mice (12–15 months old) were fed chow, Western diet (WD), WD with 4F, and WD with scramble peptide for 12 weeks. Serial echocardiography, cardiac catheterization, oxidized LDL assay, real‐time quantitative reverse transcription–polymerase chain reaction, and histological analysis were performed. The effect of LDL‐R knockdown and oxidized LDL on human pulmonary artery smooth muscle cell proliferation was assessed in vitro. LDL‐R and CD36 expression levels were significantly downregulated in the lungs of patients with PH. Patients with PH also had increased lung lipid deposits, oxidized LDL, E06 immunoreactivity, and plasma oxidized LDL/LDL ratio. LDL‐R knockout mice on WD developed PH, right ventricular hypertrophy, right ventricular dysfunction, pulmonary vascular remodeling, fibrosis, and lipid deposition in lungs, aortic atherosclerosis, and left ventricular dysfunction, which were prevented by 4F. Interestingly, PH in WD group preceded left ventricular dysfunction. Oxidized LDL or LDL‐R knockdown significantly increased proliferation of human pulmonary artery smooth muscle cells in vitro. Conclusions Human PH is associated with decreased LDL‐R in lungs and increased oxidized LDL in lungs and plasma. WD‐fed LDL‐R knockout mice develop PH and right ventricular dysfunction, implicating a role for LDL‐R and oxidized lipids in PH.
Collapse
Affiliation(s)
- Soban Umar
- Department of Anesthesiology David Geffen School of Medicine at UCLA Los Angeles CA
| | - Gregoire Ruffenach
- Department of Anesthesiology David Geffen School of Medicine at UCLA Los Angeles CA
| | - Shayan Moazeni
- Department of Anesthesiology David Geffen School of Medicine at UCLA Los Angeles CA
| | - Mylene Vaillancourt
- Department of Anesthesiology David Geffen School of Medicine at UCLA Los Angeles CA
| | - Jason Hong
- Department of Anesthesiology David Geffen School of Medicine at UCLA Los Angeles CA
| | - Christine Cunningham
- Department of Anesthesiology David Geffen School of Medicine at UCLA Los Angeles CA
| | - Nancy Cao
- Department of Anesthesiology David Geffen School of Medicine at UCLA Los Angeles CA
| | - Sara Navab
- Department of Anesthesiology David Geffen School of Medicine at UCLA Los Angeles CA
| | - Shervin Sarji
- Department of Anesthesiology David Geffen School of Medicine at UCLA Los Angeles CA
| | - Min Li
- Department of Anesthesiology David Geffen School of Medicine at UCLA Los Angeles CA
| | - Lisa Lee
- Department of Anesthesiology David Geffen School of Medicine at UCLA Los Angeles CA
| | - Greg Fishbein
- Department of Pathology David Geffen School of Medicine at UCLA Los Angeles CA
| | - Abbas Ardehali
- Department of Surgery David Geffen School of Medicine at UCLA Los Angeles CA
| | - Mohamad Navab
- Department of Medicine David Geffen School of Medicine at UCLA Los Angeles CA
| | - Srinivasa T Reddy
- Department of Medicine David Geffen School of Medicine at UCLA Los Angeles CA
| | - Mansoureh Eghbali
- Department of Anesthesiology David Geffen School of Medicine at UCLA Los Angeles CA
| |
Collapse
|
37
|
de Oliveira Junior WV, Silva APF, de Figueiredo RC, Gomes KB, Simões E Silva AC, Dusse LMS, Rios DRA. Association between dyslipidemia and CCL2 in patients undergoing hemodialysis. Cytokine 2019; 125:154858. [PMID: 31557637 DOI: 10.1016/j.cyto.2019.154858] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Revised: 08/29/2019] [Accepted: 09/12/2019] [Indexed: 12/18/2022]
Abstract
INTRODUCTION Hemodialysis (HD) is associated with high risk for cardiovascular diseases including acute myocardial infarction, stroke and congestive heart failure. C-C Motif Chemokine Ligand 2 (CCL2), also known monocyte chemotactic protein-1 (MCP-1) can be produced by a variety of cells, reaching increased levels in dyslipidemic chronic kidney disease (CKD) patients undergoing HD treatment. The main of this study was to evaluate the association between of CCL2 plasma levels and dyslipidemia in CKD patients undergoing HD. METHODS A cross-sectional study enrolled 160 Brazilian HD patients. CCL2 plasma levels were measured by capture ELISA. The association between CCL2 levels and dyslipidemia was investigated using linear regression, adjusted for classic and non-classical CVD risk factors. RESULTS A significant association was observed between CCL2 levels and dyslipidemia (P = 0.029), even after adjustment for possible confounding variables, such as age, gender, body mass index, diabetes mellitus, HD time, urea pre-hemodialysis and interdialytic weight gain (P = 0.045). CONCLUSION Our findings show that CCL2 levels are associated with dyslipidemia, which suggests a role of this cytokine in the pathogenesis of cardiovascular disease in HD patients. A better understanding of this pathogenesis could contribute to the discovery of new therapeutic targets that would reduce cardiovascular complications in these patients.
Collapse
Affiliation(s)
| | | | | | - Karina Braga Gomes
- Departamento de Análises Clínicas e Toxicológicas, Faculdade de Farmácia - Universidade Federal de Minas Gerais, Brazil
| | - Ana Cristina Simões E Silva
- Departamento de Pediatria, Laboratório Interdisciplinar de Investigação Médica, Faculdade de Medicina - Universidade Federal de Minas Gerais, Brazil
| | - Luci Maria Sant'Ana Dusse
- Departamento de Análises Clínicas e Toxicológicas, Faculdade de Farmácia - Universidade Federal de Minas Gerais, Brazil
| | | |
Collapse
|
38
|
Van den Hof M, Klein Haneveld MJ, Blokhuis C, Scherpbier HJ, Jansen HPG, Kootstra NA, Dallinga-Thie GM, Van Deventer SJH, Tsimikas S, Pajkrt D. Elevated Lipoprotein(a) in Perinatally HIV-Infected Children Compared With Healthy Ethnicity-Matched Controls. Open Forum Infect Dis 2019; 6:ofz301. [PMID: 31660394 PMCID: PMC6736182 DOI: 10.1093/ofid/ofz301] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2019] [Indexed: 01/06/2023] Open
Abstract
Background HIV-associated cardiovascular disease (CVD) risk in combination antiretroviral therapy (cART)-treated perinatally HIV-infected patients (PHIV+) remains unknown due to the young age of this population. Lipoprotein(a) (Lp(a)) has been established as an independent causal risk factor for CVD in the general population but has not been well established in the population of PHIV+. Methods We cross-sectionally compared lipid profiles, including nonfasting Lp(a), together with total cholesterol, high-density lipoprotein cholesterol, low-density lipoprotein cholesterol, and triglycerides between 35 cART-treated PHIV+ children aged 8-18 years and 37 controls who were matched for age, sex, ethnicity, and socioeconomic status. We explored associations between Lp(a) and disease- and treatment-related factors (inflammation, monocyte activation, and vascular), biomarkers, and neuroimaging outcomes using linear regression models. Results PHIV+ children had significantly higher levels of Lp(a) compared with controls (median, 43.6 [21.6-82.4] vs 21.8 [16.8-46.6] mg/dL; P = .033). Other lipid levels were comparable between groups. Additional assessment of apolipoprotein B, apolipoprotein CIII, apolipoprotein E, and APOE genotype revealed no significant differences. Higher Lp(a) levels were associated with higher plasma apoB levels and with lower monocyte chemoattractant protein-1 and TG levels in PHIV+ children. Lp(a) was not associated with HIV- or cART-related variables or with neuroimaging outcomes. Conclusions cART-treated PHIV+ children appear to have higher levels of Lp(a) compared with ethnicity-matched controls, which may implicate higher CVD risk in this population. Future research should focus on the association between Lp(a) and (sub)clinical CVD measurements in cART-treated PHIV+ patients. Dutch Trial Register number NRT4074.
Collapse
Affiliation(s)
- Malon Van den Hof
- Pediatric Infectious Diseases, Emma Children's Hospital, Amsterdam, the Netherlands
| | | | - Charlotte Blokhuis
- Pediatric Infectious Diseases, Emma Children's Hospital, Amsterdam, the Netherlands
| | | | - Hans P G Jansen
- Department of Experimental Vascular Medicine and Vascular Medicine, Amsterdam, the Netherlands
| | - Neeltje A Kootstra
- Department of Experimental Immunology, Amsterdam University Medical Centers, location Academic Medical Center, Amsterdam, the Netherlands
| | - Geesje M Dallinga-Thie
- Department of Experimental Vascular Medicine and Vascular Medicine, Amsterdam, the Netherlands
| | | | - Sotirios Tsimikas
- Sulpizio Cardiovascular Center, Division of Cardiovascular Medicine, University California San Diego, La Jolla, California
| | - Dasja Pajkrt
- Pediatric Infectious Diseases, Emma Children's Hospital, Amsterdam, the Netherlands
| | | |
Collapse
|
39
|
Coupled Modeling of Lipid Deposition, Inflammatory Response and Intraplaque Angiogenesis in Atherosclerotic Plaque. Ann Biomed Eng 2018; 47:439-452. [PMID: 30488310 DOI: 10.1007/s10439-018-02173-1] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2018] [Accepted: 11/20/2018] [Indexed: 12/13/2022]
Abstract
We propose a multiphysical mathematical model by fully coupling lipid deposition, monocytes/macrophages recruitment and angiogenesis to investigate the pathophysiological responses of an atherosclerotic plaque to the dynamic changes in the microenvironment. The time evolutions of cellular (endothelial cells, macrophages, smooth muscle cells, etc.) and acellular components (low density lipoprotein, proinflammatory cytokines, extravascular plasma concentration, etc.) within the plaque microenvironment are assessed quantitatively. The thickening of the intima, the distributions of the lipid and inflammatory factors, and the intraplaque hemorrhage show a qualitative consistency with the MRI and histology data. Models with and without angiogenesis are compared to demonstrate the important role of neovasculature in the accumulation of blood-borne components in the atherosclerotic lesion by extravasation from the leaky vessel wall, leading to the formation of a lipid core and an inflammatory microenvironment, which eventually promotes plaque destabilization. This model can serve as a theoretical platform for the investigation of the pathological mechanisms of plaque progression and may contribute to the optimal design of atherosclerosis treatment strategies, such as lipid-lowering or anti-angiogenetic therapies.
Collapse
|
40
|
Willeit P, Ridker PM, Nestel PJ, Simes J, Tonkin AM, Pedersen TR, Schwartz GG, Olsson AG, Colhoun HM, Kronenberg F, Drechsler C, Wanner C, Mora S, Lesogor A, Tsimikas S. Baseline and on-statin treatment lipoprotein(a) levels for prediction of cardiovascular events: individual patient-data meta-analysis of statin outcome trials. Lancet 2018; 392:1311-1320. [PMID: 30293769 DOI: 10.1016/s0140-6736(18)31652-0] [Citation(s) in RCA: 359] [Impact Index Per Article: 51.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/17/2018] [Revised: 07/10/2018] [Accepted: 07/12/2018] [Indexed: 12/14/2022]
Abstract
BACKGROUND Elevated lipoprotein(a) is a genetic risk factor for cardiovascular disease in general population studies. However, its contribution to risk for cardiovascular events in patients with established cardiovascular disease or on statin therapy is uncertain. METHODS Patient-level data from seven randomised, placebo-controlled, statin outcomes trials were collated and harmonised to calculate hazard ratios (HRs) for cardiovascular events, defined as fatal or non-fatal coronary heart disease, stroke, or revascularisation procedures. HRs for cardiovascular events were estimated within each trial across predefined lipoprotein(a) groups (15 to <30 mg/dL, 30 to <50 mg/dL, and ≥50 mg/dL, vs <15 mg/dL), before pooling estimates using multivariate random-effects meta-analysis. FINDINGS Analyses included data for 29 069 patients with repeat lipoprotein(a) measurements (mean age 62 years [SD 8]; 8064 [28%] women; 5751 events during 95 576 person-years at risk). Initiation of statin therapy reduced LDL cholesterol (mean change -39% [95% CI -43 to -35]) without a significant change in lipoprotein(a). Associations of baseline and on-statin treatment lipoprotein(a) with cardiovascular disease risk were approximately linear, with increased risk at lipoprotein(a) values of 30 mg/dL or greater for baseline lipoprotein(a) and 50 mg/dL or greater for on-statin lipoprotein(a). For baseline lipoprotein(a), HRs adjusted for age and sex (vs <15 mg/dL) were 1·04 (95% CI 0·91-1·18) for 15 mg/dL to less than 30 mg/dL, 1·11 (1·00-1·22) for 30 mg/dL to less than 50 mg/dL, and 1·31 (1·08-1·58) for 50 mg/dL or higher; respective HRs for on-statin lipoprotein(a) were 0·94 (0·81-1·10), 1·06 (0·94-1·21), and 1·43 (1·15-1·76). HRs were almost identical after further adjustment for previous cardiovascular disease, diabetes, smoking, systolic blood pressure, LDL cholesterol, and HDL cholesterol. The association of on-statin lipoprotein(a) with cardiovascular disease risk was stronger than for on-placebo lipoprotein(a) (interaction p=0·010) and was more pronounced at younger ages (interaction p=0·008) without effect-modification by any other patient-level or study-level characteristics. INTERPRETATION In this individual-patient data meta-analysis of statin-treated patients, elevated baseline and on-statin lipoprotein(a) showed an independent approximately linear relation with cardiovascular disease risk. This study provides a rationale for testing the lipoprotein(a) lowering hypothesis in cardiovascular disease outcomes trials. FUNDING Novartis Pharma AG.
Collapse
Affiliation(s)
- Peter Willeit
- Department of Neurology, Medical University of Innsbruck, Innsbruck, Austria; Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK.
| | - Paul M Ridker
- Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Paul J Nestel
- Baker Heart and Diabetes Institute, Melbourne, Vic, Australia
| | - John Simes
- NHMRC Clinical Trials Centre, University of Sydney, Sydney, NSW, Australia
| | - Andrew M Tonkin
- Department of Epidemiology and Preventive Medicine, Monash University, Melbourne, Vic, Australia
| | - Terje R Pedersen
- Oslo University Hospital, Ullevål, and Medical Faculty, University of Oslo, Oslo, Norway
| | - Gregory G Schwartz
- Division of Cardiology, VA Medical Center and University of Colorado School of Medicine, Denver, CO, USA
| | - Anders G Olsson
- Department of Medicine and Care, Faculty of Health Sciences, University of Linköping, Linköping, Sweden
| | - Helen M Colhoun
- MRC Human Genetics Unit, Centre for Genomic and Experimental Medicine, MRC Institute of Genetics & Molecular Medicine, Edinburgh, UK
| | - Florian Kronenberg
- Division of Genetic Epidemiology, Department of Medical Genetics, Molecular and Clinical Pharmacology, Medical University of Innsbruck, Innsbruck, Austria
| | - Christiane Drechsler
- Division of Nephrology, Department of Internal Medicine 1 and Comprehensive Heart Failure Centre, University Hospital of Würzburg, Würzburg, Germany
| | - Christoph Wanner
- Division of Nephrology, Department of Internal Medicine 1 and Comprehensive Heart Failure Centre, University Hospital of Würzburg, Würzburg, Germany
| | - Samia Mora
- Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | | | - Sotirios Tsimikas
- Vascular Medicine Program, Sulpizio Cardiovascular Center, Division of Cardiology, Department of Medicine, University of California San Diego, La Jolla, CA, USA.
| |
Collapse
|
41
|
Laina A, Gatsiou A, Georgiopoulos G, Stamatelopoulos K, Stellos K. RNA Therapeutics in Cardiovascular Precision Medicine. Front Physiol 2018; 9:953. [PMID: 30090066 PMCID: PMC6068259 DOI: 10.3389/fphys.2018.00953] [Citation(s) in RCA: 56] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2018] [Accepted: 06/28/2018] [Indexed: 12/20/2022] Open
Abstract
Since our knowledge on structure and function of messenger RNA (mRNA) has expanded from merely being an intermediate molecule between DNA and proteins to the notion that RNA is a dynamic gene regulator that can be modified and edited, RNA has become a focus of interest into developing novel therapeutic schemes. Therapeutic modulation of RNA molecules by DNA- and RNA-based therapies has broadened the scope of therapeutic targets in infectious diseases, cancer, neurodegenerative diseases and most recently in cardiovascular diseases as well. Currently, antisense oligonucleotides (ASO), small interfering RNAs (siRNAs), and microRNAs are the most widely applied therapeutic strategies to target RNA molecules and regulate gene expression and protein production. However, a number of barriers have to be overcome including instability, inadequate binding affinity and delivery to the tissues, immunogenicity, and off-target toxicity in order for these agents to evolve into efficient drugs. As cardiovascular diseases remain the leading cause of mortality worldwide, a large number of clinical trials are under development investigating the safety and efficacy of RNA therapeutics in clinical conditions such as familial hypercholesterolemia, diabetes mellitus, hypertriglyceridemia, cardiac amyloidosis, and atrial fibrillation. In this review, we summarize the clinical trials of RNA-targeting therapies in cardiovascular disease and critically discuss the advances, the outcomes, the limitations and the future directions of RNA therapeutics in precision transcriptomic medicine.
Collapse
Affiliation(s)
- Ageliki Laina
- Department of Clinical Therapeutics, Alexandra Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Aikaterini Gatsiou
- Center of Molecular Medicine, Institute of Cardiovascular Regeneration, Goethe University Frankfurt, Frankfurt, Germany.,Department of Cardiology, Center of Internal Medicine, Goethe University Frankfurt, Frankfurt, Germany.,German Center of Cardiovascular Research, Rhein-Main Partner Site, Frankfurt, Germany
| | - Georgios Georgiopoulos
- Department of Clinical Therapeutics, Alexandra Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Kimon Stamatelopoulos
- Department of Clinical Therapeutics, Alexandra Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Konstantinos Stellos
- Center of Molecular Medicine, Institute of Cardiovascular Regeneration, Goethe University Frankfurt, Frankfurt, Germany.,Department of Cardiology, Center of Internal Medicine, Goethe University Frankfurt, Frankfurt, Germany.,German Center of Cardiovascular Research, Rhein-Main Partner Site, Frankfurt, Germany.,Cardiovascular Research Centre, Institute of Genetic Medicine, Newcastle University, Newcastle upon Tyne, United Kingdom.,Department of Cardiology, Freeman Hospital, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, United Kingdom
| |
Collapse
|
42
|
Kubota Y, Folsom AR, Ballantyne CM, Tang W. Lipoprotein(a) and abdominal aortic aneurysm risk: The Atherosclerosis Risk in Communities study. Atherosclerosis 2018; 268:63-67. [PMID: 29182987 PMCID: PMC5788200 DOI: 10.1016/j.atherosclerosis.2017.10.017] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/05/2017] [Revised: 10/06/2017] [Accepted: 10/18/2017] [Indexed: 12/21/2022]
Abstract
BACKGROUND AND AIMS No prospective study has investigated whether elevated lipoprotein(a) concentrations are associated with an increased risk of abdominal aortic aneurysm (AAA). We aimed to prospectively investigate this association. METHODS In 1987-1989, the Atherosclerosis Risk in Communities study measured plasma lipoprotein(a) in 13,683 participants aged 45-64 years, without a history of AAA surgery. We followed them for incident, clinical AAA events through 2011. RESULTS During the 272,914 person-years of follow-up, over a median of 22.6 years, we documented 505 incident AAA events. The age-, sex-, and race-adjusted model showed that individuals in the highest quintile of plasma lipoprotein(a) had an increased risk of AAA. Further adjustment for the other potential confounding factors, including other plasma lipids (high- and low-density lipoprotein cholesterol and triglyceride concentrations), attenuated the association, but individuals in the highest versus lowest quintile of plasma lipoprotein(a) still had a significantly increased risk of AAA [hazard ratio (95% confidence interval): 1.57 (1.19-2.08)]. Interaction testing suggested no difference in the associations for whites and African Americans (p for interaction = 0.96). A restricted cubic spline analysis demonstrated a positive dose-response relation of plasma lipoprotein(a) with AAA, with a steep increase in AAA risk above the 75th percentile (p for overall association = 0.0086, p for non-linear association = 0.097). CONCLUSIONS In this population-based cohort study, elevated lipoprotein(a) concentrations were independently associated with an increased risk of AAA. The association reflected a threshold of increased AAA risk at high lipoprotein(a) concentrations, rather than a steady monotonic association.
Collapse
Affiliation(s)
- Yasuhiko Kubota
- Division of Epidemiology and Community Health, School of Public Health, University of Minnesota, Minneapolis, MN, USA.
| | - Aaron R Folsom
- Division of Epidemiology and Community Health, School of Public Health, University of Minnesota, Minneapolis, MN, USA
| | - Christie M Ballantyne
- Section of Cardiovascular Research, Department of Medicine, Baylor College of Medicine, Houston, TX, USA; Houston Methodist Debakey Heart and Vascular Center, Houston, TX, USA
| | - Weihong Tang
- Division of Epidemiology and Community Health, School of Public Health, University of Minnesota, Minneapolis, MN, USA
| |
Collapse
|
43
|
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: 5.9] [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]
|
44
|
Chen H, Chen L, Liang R, Wei J. Ultrasound and magnetic resonance molecular imaging of atherosclerotic neovasculature with perfluorocarbon magnetic nanocapsules targeted against vascular endothelial growth factor receptor 2 in rats. Mol Med Rep 2017; 16:5986-5996. [PMID: 28849045 PMCID: PMC5865790 DOI: 10.3892/mmr.2017.7314] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2016] [Accepted: 06/15/2017] [Indexed: 12/26/2022] Open
Abstract
The aim of the present study was to investigate the feasibility of using ultrasonography (US) and magnetic resonance (MR) for bimodal molecular imaging of atherosclerotic neovasculature with liquid perfluorocarbon magnetic nanocapsules (NCs) targeted to vascular endothelial growth factor receptor 2 (VEGFR-2). By incorporating perfluorooctyl bromide (PFOB) and superparamagnetic iron oxide (SPIO) into polylactic acid, a SPIO-embedded PFOB NC was constructed; subsequently, a VEGFR-2-targeted NC (VTNC) containing dual detectable probes was created by covalently linking a VEGFR-2 antibody onto the surface of the SPIO-embedded PFOB NC. Target specificity was verified in vitro by incubating VTNC with VEGFR-2+ or VEGFR-2− endothelial cells. Rats with vulnerable plaques were assigned to receive either an injection of VTNC (Targeted group; n=8) or an injection of NC (Nontargeted group; n=8); control rats also received an injection of VTNC (Control group; n=8). US and MR imaging of the abdominal aorta were performed to detect VTNC by measuring of the ultrasonic grayscale intensity (GSI) and MR contrast-to-noise ratio (CNR) prior to and at successive time points following VTNC and NC injections. The percent positive area (PPA) of CD31+ (PPACD31+) or VEGFR-2+ (PPAVEGFR-2+) expression was quantified by immunohistochemical staining. CD31 was used to verify the existence of endothelial cells as it is widely expressed on the surface of endothelial cells whether activated or not. The results demonstrated that VTNC was able to highly and selectively detect VEGFR-2+ endothelial cells, and GSI, CNR, PPACD31+ and PPAVEGFR-2+ were significantly increased in the targeted group compared with the nontargeted and control groups. In the control group, no atherosclerotic plaques or angiogenesis was identified, thus no expression of PPACD31+ and PPAVEGFR-2 (data not shown). There were strong correlations among GSI, CNR, PPACD31+ and PPAVEGFR-2+. In conclusion, two-probe VTNC is feasible for bimodal US and MR molecular imaging of atherosclerotic neovasculature, which may offer complementary information for the more reliable prediction of plaque vulnerability.
Collapse
Affiliation(s)
- Hua Chen
- Department of Cardiology, Fujian Medical University Union Hospital, Fujian Institute of Coronary Heart Disease, Fuzhou, Fujian 350001, P.R. China
| | - Lianglong Chen
- Department of Cardiology, Fujian Medical University Union Hospital, Fujian Institute of Coronary Heart Disease, Fuzhou, Fujian 350001, P.R. China
| | - Rongxi Liang
- Department of Ultrasonography, Fujian Medical University Union Hospital, Fuzhou, Fujian 350001, P.R. China
| | - Jin Wei
- Department of Imaging, Fujian Medical University Union Hospital, Fuzhou, Fujian 350001, P.R. China
| |
Collapse
|
45
|
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: 686] [Impact Index Per Article: 85.8] [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
|
46
|
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: 3.4] [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]
|
47
|
Pirro M, Bianconi V, Paciullo F, Mannarino MR, Bagaglia F, Sahebkar A. Lipoprotein(a) and inflammation: A dangerous duet leading to endothelial loss of integrity. Pharmacol Res 2017; 119:178-187. [DOI: 10.1016/j.phrs.2017.02.001] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/23/2016] [Revised: 01/08/2017] [Accepted: 02/02/2017] [Indexed: 12/15/2022]
|
48
|
Abstract
Lipoprotein (a) (Lp(a)) is a modified low-density lipoprotein (LDL) particle with an additional specific apolipoprotein (a), covalently attached to apolipoprotein B‑100 of LDL by a single thioester bond. Increased plasma Lp(a) level is a genetically determined, independent, causal risk factor for cardiovascular disease. The precise quantification of Lp(a) in plasma is still hampered by mass-sensitive assays, large particle variation, poor standardization and lack of assay comparability. The physiological functions of Lp(a) include wound healing, promoting tissue repair and vascular remodeling. Similarly to other lipoproteins, Lp(a) is also susceptible for oxidative modifications, leading to extensive formation of pro-inflammatory and pro-atherogenic oxidized phospholipids, oxysterols, oxidized lipid-protein adducts in Lp(a) particles, that perpetuate atherosclerotic lesion progression and intima-media thickening through induction of M1-macrophages, inflammation, autoimmunity and apoptosis. The oxidation-specific epitopes of modified lipoproteins are major targets of pre-immune, natural IgM antibodies, that may attenuate the pro-inflammatory and pro-atherogenic effects of Lp(a). Although the data are still insufficient, recent studies suggest a potential anti-neoplastic role of Lp(a).
Collapse
Affiliation(s)
- Evelyn Orsó
- Institute for Clinical Chemistry and Laboratory Medicine, University Hospital of Regensburg, Franz-Josef-Strauss-Allee 11, 93053, Regensburg, Germany
| | - Gerd Schmitz
- Institute for Clinical Chemistry and Laboratory Medicine, University Hospital of Regensburg, Franz-Josef-Strauss-Allee 11, 93053, Regensburg, Germany.
| |
Collapse
|
49
|
Byun YS, Yang X, Bao W, DeMicco D, Laskey R, Witztum JL, Tsimikas S. Oxidized Phospholipids on Apolipoprotein B-100 and Recurrent Ischemic Events Following Stroke or Transient Ischemic Attack. J Am Coll Cardiol 2017; 69:147-158. [DOI: 10.1016/j.jacc.2016.10.057] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/11/2016] [Revised: 09/29/2016] [Accepted: 10/12/2016] [Indexed: 01/08/2023]
|
50
|
Zhang X, Zhu Y, Song F, Yao Y, Ya F, Li D, Ling W, Yang Y. Effects of purified anthocyanin supplementation on platelet chemokines in hypocholesterolemic individuals: a randomized controlled trial. Nutr Metab (Lond) 2016; 13:86. [PMID: 27933092 PMCID: PMC5124283 DOI: 10.1186/s12986-016-0146-2] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2016] [Accepted: 11/17/2016] [Indexed: 12/15/2022] Open
Abstract
Background It is becoming increasingly evident that platelet chemokines are involved in distinct aspects of atherosclerosis. The aim of this study was to examine the effects of long-term supplementation with purified anthocyanins on platelet chemokines in hypercholesterolemic individuals and to identify correlations of decreased platelet chemokine levels with serum lipid and inflammatory marker levels. Methods A total of 146 hypercholesterolemic individuals were recruited and treated with 320 mg of purified anthocyanins (n = 73) or a placebo (n = 73) daily for 24 weeks in this randomized, double-blind, placebo-controlled trial. Results Anthocyanin supplementation for 24 weeks significantly decreased the plasma CXCL7 (–12.32% vs. 4.22%, P = 0.001), CXCL5 (–9.95% vs. 1.93%, P = 0.011), CXCL8 (–6.07% vs. 0.66%, P = 0.004), CXCL12 (–8.11% vs. 5.43%, P = 0.023) and CCL2 levels (–11.63% vs. 12.84%, P = 0.001) compared with the placebo. Interestingly, the decreases in the CXCL7 and CCL2 levels were both positively correlated with the decreases in the serum low-density lipoprotein-cholesterol (LDL-C), high-sensitivity C-reactive protein (hsCRP) and interleukin-1β (IL-1β) levels after anthocyanin supplementation for 24 weeks. The decrease in the CXCL8 level was negatively correlated with the increase in the how-density lipoprotein-cholesterol (HDL-C) level and was positively correlated with the decrease in the soluble P-selectin (sP-selectin) level in the anthocyanin group. In addition, a positive correlation was observed between the decreases in the CXCL12 and tumornecrosis factor-α (TNF-α) levels after anthocyanin supplementation. However, the plasma CXCL4L1, CXCL1, macrophage migration inhibitory factor (MIF) and human plasminogen activator inhibitor 1 (PAI-1) levels did not significantly change following anthocyanin supplementation. Conclusions The present study supports the notion that platelet chemokines are promising targets of anthocyanins in the prevention of atherosclerosis. Trial registration ChiCTR-TRC-08000240. Registered: 10 December 2008.
Collapse
Affiliation(s)
- Xiandan Zhang
- Department of Nutrition, School of Public Health, Sun Yat-sen University (Northern Campus), Guangzhou, People's Republic of China.,Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Guangzhou, People's Republic of China
| | - Yanna Zhu
- Department of Maternal and Child Health, School of Public Health, Sun Yat-sen University (Northern Campus), Guangzhou, People's Republic of China
| | - Fenglin Song
- School of Food Science, Guangdong Pharmaceutical University, Guangzhou, People's Republic of China
| | - Yanling Yao
- Department of Nutrition, School of Public Health, Sun Yat-sen University (Northern Campus), Guangzhou, People's Republic of China.,Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Guangzhou, People's Republic of China
| | - Fuli Ya
- Department of Nutrition, School of Public Health, Sun Yat-sen University (Northern Campus), Guangzhou, People's Republic of China.,Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Guangzhou, People's Republic of China
| | - Dan Li
- Department of Nutrition, School of Public Health, Sun Yat-sen University (Northern Campus), Guangzhou, People's Republic of China.,Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Guangzhou, People's Republic of China
| | - Wenhua Ling
- Department of Nutrition, School of Public Health, Sun Yat-sen University (Northern Campus), Guangzhou, People's Republic of China.,Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Guangzhou, People's Republic of China
| | - Yan Yang
- Department of Nutrition, School of Public Health, Sun Yat-sen University (Northern Campus), Guangzhou, People's Republic of China.,Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Guangzhou, People's Republic of China
| |
Collapse
|