1
|
Yang L, Zhang J, Han J, Jiang X. Relationship between lipoprotein B and the severity of coronary microvascular dysfunction. Clin Exp Hypertens 2025; 47:2477651. [PMID: 40102057 DOI: 10.1080/10641963.2025.2477651] [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: 01/06/2025] [Revised: 02/28/2025] [Accepted: 03/04/2025] [Indexed: 03/20/2025]
Abstract
OBJECTIVE Contributing factors for the development of heart failure (HF) involve both apolipoprotein B (ApoB) and coronary microvascular dysfunction (CMD). Although ApoB has been linked to diverse cardiovascular risks, its association with CMD remains unclear. METHODS A total of 145 patients undergoing cardiac single-photon emission computed tomography (SPECT) scan was enrolled into this retrospective study. Based on ApoB serum level, all subjects were classified into three groups (Group 1-3). Myocardial flow reserve (MFR) was calculated using myocardial blood flow (MBF) tested in different contexts. RESULTS ApoB serum level was positively correlated to rest MBF but inversely associated with stress MBF and MFR. Following adjustment for covariates, a significant relationship was observed between increased ApoB and decreased MFR. The predictive value of ApoB was test by Receiver Operating Characteristic Curve (ROC) analysis, showing an area under curve (AUC) of 0.87. CONCLUSION The findings indicated that a higher level of ApoB correlated with the severity of CMD.
Collapse
Affiliation(s)
- Lili Yang
- Department of Cardiology, Daping Hospital, The Third Military Medical University (Army Medical University), Chongqing, P.R. China
| | - Jingjing Zhang
- Department of Cardiology, Daping Hospital, The Third Military Medical University (Army Medical University), Chongqing, P.R. China
| | - Jiangyan Han
- Department of Cardiology, Daping Hospital, The Third Military Medical University (Army Medical University), Chongqing, P.R. China
| | - Xiaojuan Jiang
- Department of Cardiology, Daping Hospital, The Third Military Medical University (Army Medical University), Chongqing, P.R. China
| |
Collapse
|
2
|
Kronenberg F, Bedlington N, Ademi Z, Geantă M, Silberzahn T, Rijken M, Kaal A, Harada-Shiba M, Chen Z, Thanassoulis G, Eliasen B, Eiselé JL, Wiegman A, Ballantyne CM, Broome E, Calabrò M, Corral P, Dol A, Donato LJ, Evans E, Funabashi S, Gouni-Berthold I, Ibarluzea IG, Johnson N, Lane J, Mora S, Nordestgaard BG, Pećin I, Kaal-Poppelaars R, Langlois MR, Ray KK, Rodenbach A, Santos RD, Stroes ESG, Tada H, Vrablík M, Winokur M, Yoshida M, Nicholls SJ, Daccord M. The Brussels International Declaration on Lipoprotein(a) Testing and Management. Atherosclerosis 2025; 406:119218. [PMID: 40340180 DOI: 10.1016/j.atherosclerosis.2025.119218] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/14/2025] [Revised: 04/23/2025] [Accepted: 04/23/2025] [Indexed: 05/10/2025]
Abstract
There is striking evidence that a high lipoprotein(a) [Lp(a)] concentration is a strong, independent, and causal cardiovascular risk factor. However, Lp(a) testing rates are very low (1 %-2 %) despite the fact that 1 in 5 individuals have elevated Lp(a) concentrations. The Brussels International Declaration on Lp(a) Testing and Management was co-created by the Lp(a) International Task Force and global leaders at the Lp(a) Global Summit, held in Brussels, Belgium, on March 24-25, 2025. The event, organized by FH Europe Foundation, brought together scientific experts, people with the lived experience of elevated Lp(a) and policy makers from the European Institutions and World Health Organization. The World Heart Federation, Global Heart Hub, and European Alliance for Cardiovascular Health and scientific organizations such as European Atherosclerosis Society, and International Atherosclerosis Society were formal partners. The Summit was hosted by a Member of the European Parliament, Romana Jerković, and held under the patronage of the Polish presidency of the Council of the European Union. The Declaration calls for 1) integration of Lp(a) testing and management into Global, European and National Cardiovascular Health Plans; 2) appropriate investment, policy and programmes in targeting Lp(a) testing and management based on a recent study demonstrating the substantial overall cost-saving to health systems across the globe; 3) political commitment to mandate systematic Lp(a) testing at least once during a person's lifetime, ideally at an early age, with full reimbursement; 4) incorporation of Lp(a) test results in the context of a person's cardiovascular risk assessment, with development of personalised cardiovascular health roadmaps as needed, without fear of dredit aiscrimination; 5) investment in public and healthcare professional education to increase awareness of Lp(a) and its impact on cardiovascular health.
Collapse
Affiliation(s)
- Florian Kronenberg
- Institute of Genetic Epidemiology, Medical University of Innsbruck, Innsbruck, Austria; Lp(a) International Task Force, FH Europe Foundation, Amsterdam, Netherlands.
| | - Nicola Bedlington
- Lp(a) International Task Force, FH Europe Foundation, Amsterdam, Netherlands
| | - Zanfina Ademi
- Lp(a) International Task Force, FH Europe Foundation, Amsterdam, Netherlands; Health Economics and Policy Evaluation Research (HEPER) Group, Centre for Medicine Use and Safety, Faculty of Pharmacy and Pharmaceutical Sciences, Monash University, Melbourne, Australia
| | - Marius Geantă
- Lp(a) International Task Force, FH Europe Foundation, Amsterdam, Netherlands; Center for Innovation in Medicine, Bucharest, Romania
| | - Tobias Silberzahn
- Lp(a) International Task Force, FH Europe Foundation, Amsterdam, Netherlands
| | - Marc Rijken
- Lp(a) International Task Force, FH Europe Foundation, Amsterdam, Netherlands
| | - Aedan Kaal
- FH Europe Foundation, Amsterdam, Netherlands
| | - Mariko Harada-Shiba
- Lp(a) International Task Force, FH Europe Foundation, Amsterdam, Netherlands; Cardiovascular Center, Osaka Medical and Pharmaceutical University, Osaka, Japan
| | - Zhenyue Chen
- Lp(a) International Task Force, FH Europe Foundation, Amsterdam, Netherlands; Department of Cardiovascular Medicine, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - George Thanassoulis
- Lp(a) International Task Force, FH Europe Foundation, Amsterdam, Netherlands; Preventive and Genomic Cardiology, McGill University Health Center, Montréal, Québec, Canada
| | - Bogi Eliasen
- Lp(a) International Task Force, FH Europe Foundation, Amsterdam, Netherlands; Movement Health Foundation, Switzerland
| | - Jean-Luc Eiselé
- Lp(a) International Task Force, FH Europe Foundation, Amsterdam, Netherlands; World Heart Federation (WHF), Geneva, Switzerland
| | - Albert Wiegman
- Lp(a) International Task Force, FH Europe Foundation, Amsterdam, Netherlands; Department of Pediatrics Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, Netherlands
| | - Christie M Ballantyne
- Sections of Cardiology and Cardiovascular Research, Department of Medicine, Baylor College of Medicine and the Texas Heart Institute, Houston, TX, USA
| | - Emma Broome
- FH Europe Foundation, Amsterdam, Netherlands
| | - Michele Calabrò
- European Regional and Local Health Authorities, Brussels, Belgium
| | - Pablo Corral
- FASTA University, School of Medicine, Pharmacology and Research Department, Mar Del Plata, Argentina
| | | | - Leslie J Donato
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA
| | - Elsie Evans
- FH Europe Foundation, Amsterdam, Netherlands
| | - Sayaka Funabashi
- Department of Cardiovascular Medicine, Kyorin University, Faculty of Medicine, Mitaka, Japan
| | - Ioanna Gouni-Berthold
- Center for Endocrinology, Diabetes and Preventive Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | | | | | - Joanna Lane
- FH Europe Foundation, Amsterdam, Netherlands; Stichting Health Clusternet, Amsterdam, Netherlands; Centre for Health and Technology, University of South-Eastern Norway, Norway
| | - Samia Mora
- International Atherosclerosis Society, Washington, DC, USA; Center for Lipid Metabolomics, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Børge G Nordestgaard
- Department of Clinical Biochemistry, Copenhagen University Hospital - Herlev and Gentofte, Denmark; The Copenhagen General Population Study, Copenhagen University Hospital - Herlev and Gentofte, Denmark; Institute of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Denmark
| | - Ivan Pećin
- Zagreb School of Medicine, University of Zagreb, University Hospital Center Zagreb, Zagreb, Croatia
| | | | - Michel R Langlois
- EFLM (European Federation of Clinical Chemistry and Laboratory Medicine), Brussels, Belgium
| | - Kausik K Ray
- Department of Public Health and Primary Care, Imperial College London, London, UK
| | - Arthur Rodenbach
- Department of Internal Medicine and Pediatrics, Ghent University Hospital, Ghent, Belgium
| | - Raul D Santos
- Hospital Israelita Albert Einstein and University of Sao Paulo, Brazil
| | - Erik S G Stroes
- Department of Vascular Medicine, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, Netherlands
| | - Hayato Tada
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Japan
| | - Michal Vrablík
- 3rd Department of Internal Medicine, 1st Faculty of Medicine, Charles University, General University Hospital, Prague, Czech Republic
| | | | - Masayuki Yoshida
- Institute of Science Tokyo, Depatment of Medical Genetics, Tokyo, Japan
| | | | - Magdalena Daccord
- Lp(a) International Task Force, FH Europe Foundation, Amsterdam, Netherlands; FH Europe Foundation, Amsterdam, Netherlands.
| |
Collapse
|
3
|
Borén J, Packard CJ, Binder CJ. Apolipoprotein B-containing lipoproteins in atherogenesis. Nat Rev Cardiol 2025; 22:399-413. [PMID: 39743565 DOI: 10.1038/s41569-024-01111-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/25/2024] [Indexed: 01/04/2025]
Abstract
Apolipoprotein B (apoB) is the main structural protein of LDLs, triglyceride-rich lipoproteins and lipoprotein(a), and is crucial for their formation, metabolism and atherogenic properties. In this Review, we present insights into the role of apoB-containing lipoproteins in atherogenesis, with an emphasis on the mechanisms leading to plaque initiation and growth. LDL, the most abundant cholesterol-rich lipoprotein in plasma, is causally linked to atherosclerosis. LDL enters the artery wall by transcytosis and, in vulnerable regions, is retained in the subendothelial space by binding to proteoglycans via specific sites on apoB. A maladaptive response ensues. This response involves modification of LDL particles, which promotes LDL retention and the release of bioactive lipid products that trigger inflammatory responses in vascular cells, as well as adaptive immune responses. Resident and recruited macrophages take up modified LDL, leading to foam cell formation and ultimately cell death due to inadequate cellular lipid handling. Accumulation of dead cells and cholesterol crystallization are hallmarks of the necrotic core of atherosclerotic plaques. Other apoB-containing lipoproteins, although less abundant, have substantially greater atherogenicity per particle than LDL. These lipoproteins probably contribute to atherogenesis in a similar way to LDL but might also induce additional pathogenic mechanisms. Several targets for intervention to reduce the rate of atherosclerotic lesion initiation and progression have now been identified, including lowering plasma lipoprotein levels and modulating the maladaptive responses in the artery wall.
Collapse
Affiliation(s)
- Jan Borén
- Department of Molecular and Clinical Medicine, University of Gothenburg, Gothenburg, Sweden.
| | - Chris J Packard
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, UK
| | - Christoph J Binder
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
4
|
Mohammadnia N, van Broekhoven A, Bax WA, Eikelboom JW, Mosterd A, Fiolet ATL, Tijssen JGP, Thompson PL, de Kleijn DPV, Tsimikas S, Cornel JH, Yeang C, El Messaoudi S. Interleukin-6 modifies Lipoprotein(a) and oxidized phospholipids associated cardiovascular disease risk in a secondary prevention cohort. Atherosclerosis 2025; 405:119211. [PMID: 40318255 DOI: 10.1016/j.atherosclerosis.2025.119211] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Revised: 04/02/2025] [Accepted: 04/14/2025] [Indexed: 05/07/2025]
Abstract
BACKGROUND AND AIMS There is a need for effective tools to stratify and modify cardiovascular risk associated with elevated lipoprotein(a) [Lp(a)] and oxidized phospholipids (OxPL). The objective of this analysis was to explore the modifying effects of low-grade inflammation on Lp(a)- and OxPL-associated risk in a secondary prevention cohort. METHODS Levels of Lp(a), OxPL associated with apolipoprotein(a) (OxPL-apo[a]) and apolipoprotein B (OxPL-apoB) were determined in the placebo-arm of the low-dose colchicine 2 trial. Patients were between 35 and 82 years, had established chronic coronary syndrome (CCS), and were clinically stable for at least six months prior to randomization. The outcome was the incidence of the composite endpoint of spontaneous myocardial infarction, ischemic stroke, or ischemia-driven coronary revascularization stratified by biomarker levels using a Cox regression model. RESULTS There was a significant interaction between Lp(a) and IL-6 <3.2 ng/L (median) and IL-6 ≥3.2 ng/L for the composite endpoint (HR 0.90; 95 %CI 0.78-1.03 vs HR 1.18; 95 %CI 1.01-1.39, Pinteraction = 0.01). No interaction was found for Lp(a) levels in participants with hsCRP <2 mg/L (HR 1.00; 95 %CI 0.89-1.14) versus those with hsCRP ≥2 mg/L (HR 1.04; 95 %CI 0.86-1.25, Pinteraction = 0.79). In line with Lp(a) levels, significant interaction was observed between OxPL-apo(a) as well as OxPL-apoB levels for the composite endpoint with IL-6 (Pinteraction<0.01 and 0.03, respectively), but not for hsCRP. CONCLUSIONS In patients with CCS, Lp(a), OxPL-apo(a) and OxPL-apoB associated cardiovascular risk was only pertinent in those with elevated IL-6 but not hsCRP levels.
Collapse
Affiliation(s)
| | - Amber van Broekhoven
- Department of Cardiology, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Willem A Bax
- Department of Internal Medicine, Northwest Clinics, Alkmaar, the Netherlands
| | - John W Eikelboom
- Department of Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Arend Mosterd
- Dutch Network for Cardiovascular Research (WCN), Utrecht, the Netherlands; Department of Cardiology, Meander Medical Center, Amersfoort, the Netherlands
| | - Aernoud T L Fiolet
- Dutch Network for Cardiovascular Research (WCN), Utrecht, the Netherlands; Department of Cardiology, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Jan G P Tijssen
- Department of Cardiology, Amsterdam University Medical Centers, Amsterdam, the Netherlands
| | - Peter L Thompson
- Heart and Vascular Research Institute of Western Australia, Perth, Australia; School of Medicine, University of Western Australia, Perth, Australia; Sir Charles Gairdner Hospital, Perth, Australia
| | - Dominique P V de Kleijn
- The Netherlands Heart Institute, Utrecht, the Netherlands; Department of Vascular Surgery, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Sotirios Tsimikas
- Department of Medicine, University of California, La Jolla, San Diego, CA, USA
| | - Jan H Cornel
- Department of Cardiology, Radboud University Medical Center, Nijmegen, the Netherlands; Dutch Network for Cardiovascular Research (WCN), Utrecht, the Netherlands; Department of Cardiology, Northwest Clinics, Alkmaar, the Netherlands.
| | - Calvin Yeang
- Department of Medicine, University of California, La Jolla, San Diego, CA, USA.
| | - Saloua El Messaoudi
- Department of Cardiology, Radboud University Medical Center, Nijmegen, the Netherlands
| |
Collapse
|
5
|
Rosenson RS, López JAG, Gaudet D, Baum SJ, Stout E, Lepor NE, Park JG, Murphy SA, Knusel B, Wang J, Wilmanski T, Wang H, Wu Y, Kassahun H, Sabatine MS, O’Donoghue ML. Olpasiran, Oxidized Phospholipids, and Systemic Inflammatory Biomarkers: Results From the OCEAN(a)-DOSE Trial. JAMA Cardiol 2025; 10:482-486. [PMID: 39937508 PMCID: PMC11822594 DOI: 10.1001/jamacardio.2024.5433] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Accepted: 11/22/2024] [Indexed: 02/13/2025]
Abstract
Importance Lipoprotein(a) (Lp[a]) is thought to be the major carrier of oxidized phospholipids (OxPL). OxPL are believed to be a potent driver of inflammation and atherosclerosis. Olpasiran, a small interfering RNA, blocks Lp(a) production by inducing degradation of apolipoprotein(a) messenger RNA. Olpasiran's effects on OxPL and systemic markers of inflammation are not well described. Objective To assess the effects of olpasiran on OxPL, high-sensitivity interleukin 6 (hs-IL-6), and hs-C-reactive protein (hs-CRP) in the OCEAN(a)-DOSE randomized clinical trial. Design, Setting, and Participants OCEAN(a)-DOSE was an international, multicenter, placebo-controlled, phase 2, dose-finding randomized clinical trial conducted between July 2020 and November 2022. A total of 281 patients with atherosclerotic cardiovascular disease and Lp(a) levels greater than 150 nmol/L were included. Intervention Participants were randomized to receive 1 of 4 active subcutaneous doses of olpasiran vs placebo: (1) 10 mg, administered every 12 weeks (Q12W); (2) 75 mg, Q12W; (3) 225 mg, Q12W; or (4) 225 mg, administered every 24 weeks (Q24W). OxPL on apolipoprotein B (OxPL-apoB), hs-CRP, and hs-IL-6 were assessed at baseline, week 36, and week 48 in 272 patients. Main Outcomes and Measures The primary outcome was placebo-adjusted change in OxPL-apoB from baseline to week 36. Results Among 272 participants, median (IQR) age was 62 years (56-69), and 86 participants (31.6%) were female. Baseline median (IQR) Lp(a) concentration was 260.3 nmol/L (198.1-352.4) and median (IQR) OxPL-apoB concentration was 26.5 nmol/L (19.7-33.9). The placebo-adjusted mean percentage change in OxPL-apoB from baseline to week 36 was -51.6% (95% CI, -64.9% to -38.2%) for the 10-mg Q12W dose, -89.7% (95% CI, -103.0% to -76.4%) for the 75-mg Q12W dose, -92.3% (95% CI, -105.6% to -78.9%) for the 225-mg Q12W dose, and -93.7% (95% CI, -107.1% to -80.3%) for the Q24W dose (P < .001 for all). These effects were maintained to week 48 (-50.8%, -100.2%, -104.7%, and -85.8%, respectively; P < .001 for all). There was a strong correlation between percentage reduction in Lp(a) and OxPL-apoB for patients treated with olpasiran (r = 0.79; P < .001). Olpasiran did not significantly impact hs-CRP or hs-IL-6 compared with placebo to weeks 36 or 48 (P > .05). Conclusion and Relevance In the OCEAN(a)-DOSE multicenter randomized clinical trial, olpasiran led to a significant and sustained reduction in OxPL-apoB but no significant effects on hs-CRP or hs-IL-6.
Collapse
Affiliation(s)
- Robert S. Rosenson
- Metabolism and Lipids Program, Icahn School of Medicine, Mount Sinai Hospital, New York, New York
| | | | - Daniel Gaudet
- Department of Medicine, Université de Montréal, Chicoutimi, Quebec, Canada
| | - Seth J. Baum
- Flourish Research, Boca Raton, Florida
- Charles E Schmidt College of Medicine, Florida Atlantic University, Boca Raton, Florida
| | - Elmer Stout
- Crossroads Clinical Research Inc, Mooresville, North Carolina
| | - Norman E. Lepor
- David Geffen School of Medicine, University of California, Los Angeles
| | - Jeong-Gun Park
- TIMI Study Group, Division of Cardiovascular Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts
| | - Sabina A. Murphy
- TIMI Study Group, Division of Cardiovascular Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts
| | - Beat Knusel
- Global Development, Amgen, Thousand Oaks, California
| | - Jingying Wang
- Global Development, Amgen, Thousand Oaks, California
| | | | - Huei Wang
- Global Development, Amgen, Thousand Oaks, California
| | - You Wu
- Global Development, Amgen, Thousand Oaks, California
| | | | - Marc S. Sabatine
- TIMI Study Group, Division of Cardiovascular Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts
| | - Michelle L. O’Donoghue
- TIMI Study Group, Division of Cardiovascular Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
6
|
Farina CJ, Lu W, Nilsson J. Orticumab: the potential to harness oxidized LDL to reduce coronary inflammation with plaque-targeted therapy. Curr Opin Lipidol 2025:00041433-990000000-00120. [PMID: 40293233 DOI: 10.1097/mol.0000000000000990] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 04/30/2025]
Abstract
PURPOSE OF REVIEW Myocardial infarction survivors are at a high risk of a recurrent event despite receiving guideline preventive therapy. There is accumulated evidence that persistent atherosclerotic plaque inflammation contributes to this risk. Oxidized low-density lipoprotein (LDL) is widely recognized as a key factor in plaque inflammation and instability; however, no therapies that directly target oxidized LDL are to date available for clinical use. We will here review recent observations indicating that treatment with the anti-oxidized LDL antibody orticumab specifically inhibits plaque inflammation. RECENT FINDINGS The effect of orticumab on coronary inflammation in a randomized, double-blind, placebo-controlled pilot phase 2a trial in subjects with moderate to severe psoriasis is a new and recent finding. Coronary inflammation was assessed by calculation of the fat attenuation index (FAI)-Score in the pericoronary adipose tissue in coronary computed tomography angiograms. After 15 weeks of treatment the mean FAI-Score of the three main coronary arteries was significantly reduced in the orticumab group while no change occurred in the placebo group. The effect of orticumab was most pronounced in those with most inflammation at baseline. SUMMARY Treatment with orticumab represents a new and plaque-specific way to reduce arterial inflammation.
Collapse
Affiliation(s)
| | - Wenqi Lu
- Abcentra, Los Angeles, California, USA
| | - Jan Nilsson
- Abcentra, Los Angeles, California, USA
- Department of Clinical Sciences Malmö, Lund University, Lund, Sweden
| |
Collapse
|
7
|
Wang X, Xie Z, Zhang J, Chen Y, Li Q, Yang Q, Chen X, Liu B, Xu S, Dong Y. Interaction between lipid metabolism and macrophage polarization in atherosclerosis. iScience 2025; 28:112168. [PMID: 40201117 PMCID: PMC11978336 DOI: 10.1016/j.isci.2025.112168] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/10/2025] Open
Abstract
Atherosclerosis (AS) is a chronic inflammatory condition associated with lipid deposition. The interaction between abnormal lipid metabolism and the inflammatory response has been identified as the underlying cause of AS. Lipid metabolism disorders are considered the basis of atherosclerotic lesion formation and macrophages are involved in the entire process of AS formation. Macrophages have a high degree of plasticity, and the change of their polarization direction can determine the progress or regression of AS. The disturbances in bioactive lipid metabolism affect the polarization of different phenotypes of macrophages, thus, affecting lipid metabolism and the expression of key signal factors. Therefore, understanding the interaction between lipid metabolism and macrophages as well as their key targets is important for preventing and treating AS and developing new drugs. Recent studies have shown that traditional Chinese medicines play a positive role in the prevention and treatment of AS, providing a basis for clinical individualized treatment.
Collapse
Affiliation(s)
- Xinge Wang
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, China; School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
- Guang’ anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China
| | - Zheng Xie
- Guang’ anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China
| | - Jing Zhang
- Tianjin State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Ying Chen
- Institute of Chinese Materia Medica China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Qi Li
- Institute of Chinese Materia Medica China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Qing Yang
- Institute of Chinese Materia Medica China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Xu Chen
- Guang’ anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China
| | - Bing Liu
- Guang’ anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China
| | - Shijun Xu
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, China; School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Yu Dong
- Guang’ anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China
| |
Collapse
|
8
|
Tang H, Kan C, Zhang K, Sheng S, Qiu H, Ma Y, Wang Y, Hou N, Zhang J, Sun X. Glycerophospholipid and Sphingosine- 1-phosphate Metabolism in Cardiovascular Disease: Mechanisms and Therapeutic Potential. J Cardiovasc Transl Res 2025:10.1007/s12265-025-10620-3. [PMID: 40227543 DOI: 10.1007/s12265-025-10620-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Accepted: 04/07/2025] [Indexed: 04/15/2025]
Abstract
Cardiovascular disease remains a leading cause of mortality worldwide, driven by factors such as dysregulated lipid metabolism, oxidative stress, and inflammation. Recent studies highlight the critical roles of both glycerophospholipid and sphingosine- 1-phosphate metabolism in the pathogenesis of cardiovascular disorders. However, the contributions of glycerophospholipid-derived metabolites remain underappreciated. Glycerophospholipid metabolism generates bioactive molecules that contribute to endothelial dysfunction, lipid accumulation, and cardiac cell injury while also modulating inflammatory and oxidative stress responses. Meanwhile, sphingosine- 1-phosphate is a bioactive lipid mediator that regulates vascular integrity, inflammation, and cardiac remodeling through its G-protein-coupled receptors. The convergence of these pathways presents novel therapeutic opportunities, where dietary interventions such as omega- 3 polyunsaturated fatty acids and pharmacological targeting of sphingosine- 1-phosphate receptors could synergistically mitigate cardiovascular risk. This review underscores the need for further investigation into the interplay between glycerophospholipid metabolism and sphingosine- 1-phosphate signaling to advance targeted therapies for the prevention and management of cardiovascular disease.
Collapse
Affiliation(s)
- Huiru Tang
- Department of Endocrinology and Metabolism, Clinical Research Center, Shandong Provincial Key Medical and Health Discipline of Endocrinology, Affiliated Hospital of Shandong Second Medical University, Weifang, China
| | - Chengxia Kan
- Department of Endocrinology and Metabolism, Clinical Research Center, Shandong Provincial Key Medical and Health Discipline of Endocrinology, Affiliated Hospital of Shandong Second Medical University, Weifang, China
| | - Kexin Zhang
- Department of Endocrinology and Metabolism, Clinical Research Center, Shandong Provincial Key Medical and Health Discipline of Endocrinology, Affiliated Hospital of Shandong Second Medical University, Weifang, China
| | - Sufang Sheng
- Department of Endocrinology and Metabolism, Clinical Research Center, Shandong Provincial Key Medical and Health Discipline of Endocrinology, Affiliated Hospital of Shandong Second Medical University, Weifang, China
| | - Hongyan Qiu
- Department of Endocrinology and Metabolism, Clinical Research Center, Shandong Provincial Key Medical and Health Discipline of Endocrinology, Affiliated Hospital of Shandong Second Medical University, Weifang, China
| | - Yujie Ma
- Department of Endocrinology and Metabolism, Clinical Research Center, Shandong Provincial Key Medical and Health Discipline of Endocrinology, Affiliated Hospital of Shandong Second Medical University, Weifang, China
| | - Yuqun Wang
- Department of Endocrinology and Metabolism, Clinical Research Center, Shandong Provincial Key Medical and Health Discipline of Endocrinology, Affiliated Hospital of Shandong Second Medical University, Weifang, China
| | - Ningning Hou
- Department of Endocrinology and Metabolism, Clinical Research Center, Shandong Provincial Key Medical and Health Discipline of Endocrinology, Affiliated Hospital of Shandong Second Medical University, Weifang, China
| | - Jingwen Zhang
- Department of Endocrinology and Metabolism, Clinical Research Center, Shandong Provincial Key Medical and Health Discipline of Endocrinology, Affiliated Hospital of Shandong Second Medical University, Weifang, China.
| | - Xiaodong Sun
- Department of Endocrinology and Metabolism, Clinical Research Center, Shandong Provincial Key Medical and Health Discipline of Endocrinology, Affiliated Hospital of Shandong Second Medical University, Weifang, China.
| |
Collapse
|
9
|
Durrington PN, Bashir B, Soran H. How Does HDL Participate in Atherogenesis? Antioxidant Activity Versus Role in Reverse Cholesterol Transport. Antioxidants (Basel) 2025; 14:430. [PMID: 40298833 PMCID: PMC12023944 DOI: 10.3390/antiox14040430] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2025] [Revised: 03/10/2025] [Accepted: 03/15/2025] [Indexed: 04/30/2025] Open
Abstract
Low-density lipoprotein (LDL) chemically modified by reactive oxygen species (ROS), for example, leaking from red blood cells in the vascular compartment, more readily crosses the vascular endothelium than does nonoxidatively modified LDL to enter tissue fluid. Oxidatively modified LDL (oxLDL) may also be created in the tissue fluid by ROS leaking from cells by design, for example, by inflammatory white cells, or simply leaking from other cells as a consequence of oxygen metabolism. As well as oxLDL, glycatively modified LDL (glycLDL) is formed in the circulation. High-density lipoprotein (HDL) appears capable of decreasing the burden of lipid peroxides formed on LDL exposed to ROS or to glucose and its metabolites. The mechanism for this that has received the most attention is the antioxidant activity of HDL, which is due in large part to the presence of paraoxonase 1 (PON1). PON1 is intimately associated with its apolipoprotein A1 component and with HDL's lipid domains into which lipid peroxides from LDL or cell membranes can be transferred. It is frequently overlooked that for PON1 to hydrolyze lipid substrates, it is essential that it remain by virtue of its hydrophobic amino acid sequences within a lipid micellar environment, for example, during its isolation from serum or genetically modified cells in tissue culture. Otherwise, it may retain its capacity to hydrolyze water-soluble substrates, such as phenyl acetate, whilst failing to hydrolyze more lipid-soluble molecules. OxLDL and probably glycLDL, once they have crossed the arterial endothelium by receptor-mediated transcytosis, are rapidly taken up by monocytes in a process that also involves scavenger receptors, leading to subendothelial foam cell formation. These are the precursors of atheroma, inducing more monocytes to cross the endothelium into the lesion and the proliferation and migration of myocytes present in the arterial wall into the developing lesion, where they transform into foam cells and fibroblasts. The atheroma progresses to have a central extracellular lake of cholesteryl ester following necrosis and apoptosis of foam cells with an overlying fibrous cap whilst continuing to grow concentrically around the arterial wall by a process involving oxLDL and glycLDL. Within the arterial wall, additional oxLDL is generated by ROS secreted by inflammatory cells and leakage from cells generally when couplet oxygen is reduced. PON1 is important for the mechanism by which HDL opposes atherogenesis, which may provide a better avenue of inquiry in the identification of vulnerable individuals and the provision of new therapies than have emerged from the emphasis placed on its role in RCT.
Collapse
Affiliation(s)
- Paul N. Durrington
- Faculty of Biology, Medicine and Health, Cardiovascular Research Group, University of Manchester, Manchester M13 9NT, UK; (B.B.); (H.S.)
| | - Bilal Bashir
- Faculty of Biology, Medicine and Health, Cardiovascular Research Group, University of Manchester, Manchester M13 9NT, UK; (B.B.); (H.S.)
- Department of Diabetes, Endocrinology and Metabolism, Peter Mount Building, Manchester University NHS Foundation Trust, Manchester M13 9WL, UK
| | - Handrean Soran
- Faculty of Biology, Medicine and Health, Cardiovascular Research Group, University of Manchester, Manchester M13 9NT, UK; (B.B.); (H.S.)
- Department of Diabetes, Endocrinology and Metabolism, Peter Mount Building, Manchester University NHS Foundation Trust, Manchester M13 9WL, UK
| |
Collapse
|
10
|
Li Y, Yang B, Li N, Wei J, Wu Y. Association between dietary niacin intake and atherosclerotic cardiovascular disease among American adults: national health and nutrition examination survey. Front Nutr 2025; 12:1566684. [PMID: 40230721 PMCID: PMC11994425 DOI: 10.3389/fnut.2025.1566684] [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: 01/27/2025] [Accepted: 03/13/2025] [Indexed: 04/16/2025] Open
Abstract
Background The relationship between dietary niacin and atherosclerotic cardiovascular disease (ASCVD) is still not fully understood. Our objective was to assess the association between dietary niacin intake and the prevalence of ASCVD. Methods In this cross-sectional study, we examined a cohort of 15,685 adult individuals in the United States, aged 20 years and older, who participated in the National Health and Nutrition Examination Survey (NHANES) carried out between 2007 and 2014. Dietary Niacin consumption was assessed using a 24-h dietary recall method. The assessment of the presence of ASCVD was conducted through the Patient Medical Conditions Questionnaire. To assess the reliability of the results, restricted cubic spline models and logistic regression analyses were employed, along with conducting subgroup analyses. Results The analysis included 15,685 participants who were 20 years or older, drawn from the NHANES data for the cycles spanning 2007 to 2014. Of which 10.4% (1,638/15,685) were diagnosed with ASCVD. The probability of ASCVD diminishes by 9% with each 10 mg/day increment in dietary niacin intake (OR = 0.91, 95% CI: 0.87-0.96). This association held true when niacin consumption was assessed as a categorical variable. Compared to individuals with the lowest dietary niacin intake, defined as T1 (≤17.4 mg/day), the adjusted odds ratios for ASCVD in those with higher niacin intakes, T2 (17.5-27.2 mg/day) and T3 (≥27.3 mg/day), were 0.87 (95% CI: 0.76-0.99, p = 0.037) and 0.75 (95% CI: 0.64-0.87, p < 0.001), respectively. There was an inverse association between dietary niacin intake and ASCVD prevalence, supported by sensitivity analyses. Subgroup analysis revealed an interaction effect when stratified by age. Conclusion This analysis of NHANES data has demonstrated that niacin is significantly negative associated with ASCVD in American adults aged ≥20 years.
Collapse
Affiliation(s)
- Yan Li
- Department of Cardiology, Xi'an International Medical Center Hospital, Xi’an, Shaanxi, China
| | - Beilei Yang
- Department of Cardiology, Xi'an International Medical Center Hospital, Xi’an, Shaanxi, China
| | - Na Li
- Department of Cardiology, Xi'an International Medical Center Hospital, Xi’an, Shaanxi, China
| | - Jinjuan Wei
- Department of Cardiology, Xi'an International Medical Center Hospital, Xi’an, Shaanxi, China
| | - Yue Wu
- Department of Cardiovascular Medicine, People’s Hospital of Xiangxi Tujia and Miao Autonomous Prefecture, The First Affiliated Hospital of Jishou University, Jishou, China
| |
Collapse
|
11
|
Girard AS, Paulin A, Manikpurage HD, Lajeunesse E, Clavel MA, Pibarot P, Krege JH, Mathieu P, Thériault S, Arsenault BJ. Impact of Lipoprotein(a) on Valvular and Cardiovascular Outcomes in Patients With Calcific Aortic Valve Stenosis. J Am Heart Assoc 2025; 14:e038955. [PMID: 40079323 DOI: 10.1161/jaha.124.038955] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Accepted: 01/16/2025] [Indexed: 03/15/2025]
Abstract
BACKGROUND Lp(a) (lipoprotein(a)) is an independent risk factor for calcific aortic valve stenosis (CAVS). Whether patients with CAVS and high Lp(a) levels are at higher risk of valvular or cardiovascular events is unknown. The aim of this study is to determine whether higher Lp(a) levels are associated with valvular and cardiovascular outcomes in patients with CAVS. METHODS AND RESULTS We identified 1962 patients from the UK Biobank with an electronic health record or self-reported CAVS diagnosis but who did not previously undergo aortic valve replacement (AVR) and had a minimal follow-up time of 2.5 years. Cox proportional hazard regression was used to evaluate the effect of Lp(a) on AVR, AVR or cardiac death, and valvular or cardiovascular events (AVR, cardiac death, myocardial infarction, stroke, heart failure, or coronary artery bypass grafting). The maximal follow-up time was set to 5 years. During the follow-up, 198 patients underwent AVR, 260 had AVR or cardiac death, and 435 had at least 1 valvular or cardiovascular event. Patients with Lp(a) levels ≥125 versus <125 nmol/L were at higher risk of AVR (hazard ratio [HR], 1.58 [95% CI, 1.17-2.12]), AVR or cardiac death (HR, 1.43 [95% CI, 1.10-1.86]), and cardiovascular or valvular events (HR, 1.36 [95% CI, 1.11-1.68]). Point estimates were comparable in men versus women, younger versus older patients, and in patients with higher versus lower plasma C-reactive protein levels. CONCLUSIONS In patients with CAVS, Lp(a) levels predicted a higher risk of valvular and cardiovascular outcomes. The impact of Lp(a)-lowering therapies on valvular and cardiovascular health should be assessed in a long-term randomized clinical trial.
Collapse
Affiliation(s)
- Arnaud S Girard
- Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec-Université Laval Québec QC Canada
| | - Audrey Paulin
- Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec-Université Laval Québec QC Canada
| | - Hasanga D Manikpurage
- Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec-Université Laval Québec QC Canada
| | - Emma Lajeunesse
- Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec-Université Laval Québec QC Canada
| | - Marie-Annick Clavel
- Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec-Université Laval Québec QC Canada
- Department of Medicine, Faculty of Medicine Université Laval Québec QC Canada
| | - Philippe Pibarot
- Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec-Université Laval Québec QC Canada
- Department of Medicine, Faculty of Medicine Université Laval Québec QC Canada
| | | | - Patrick Mathieu
- Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec-Université Laval Québec QC Canada
- Department of Surgery, Faculty of Medicine Université Laval Québec QC Canada
| | - Sébastien Thériault
- Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec-Université Laval Québec QC Canada
- Department of Molecular Biology, Medical Biochemistry and Pathology, Faculty of Medicine Université Laval Québec QC Canada
| | - Benoit J Arsenault
- Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec-Université Laval Québec QC Canada
- Department of Medicine, Faculty of Medicine Université Laval Québec QC Canada
| |
Collapse
|
12
|
Cheng Y, Shao S, Wang Z, Guan Q, Li H, Liu G, Zhang H, Fan X, Zhao J. From lipotoxicity to pan-lipotoxicity. Cell Discov 2025; 11:27. [PMID: 40102396 PMCID: PMC11920229 DOI: 10.1038/s41421-025-00787-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/20/2025] Open
Affiliation(s)
- Yiping Cheng
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education; Department of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
- Shandong Key Laboratory of Endocrine Metabolism and Aging, Jinan, Shandong, China
- Shandong Institute of Endocrine and Metabolic Diseases, Jinan, Shandong, China
- Shandong Engineering Laboratory of Prevention and Control for Endocrine and Metabolic Diseases, Jinan, Shandong, China
| | - Shanshan Shao
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education; Department of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
- Shandong Key Laboratory of Endocrine Metabolism and Aging, Jinan, Shandong, China
- Shandong Institute of Endocrine and Metabolic Diseases, Jinan, Shandong, China
- Shandong Engineering Laboratory of Prevention and Control for Endocrine and Metabolic Diseases, Jinan, Shandong, China
| | - Zhen Wang
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education; Department of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
- Shandong Key Laboratory of Endocrine Metabolism and Aging, Jinan, Shandong, China
- Shandong Institute of Endocrine and Metabolic Diseases, Jinan, Shandong, China
- Shandong Engineering Laboratory of Prevention and Control for Endocrine and Metabolic Diseases, Jinan, Shandong, China
| | - Qingbo Guan
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education; Department of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
- Shandong Key Laboratory of Endocrine Metabolism and Aging, Jinan, Shandong, China
- Shandong Institute of Endocrine and Metabolic Diseases, Jinan, Shandong, China
- Shandong Engineering Laboratory of Prevention and Control for Endocrine and Metabolic Diseases, Jinan, Shandong, China
| | - Huaxue Li
- Shandong Key Laboratory of Endocrine Metabolism and Aging, Jinan, Shandong, China
- Shandong Institute of Endocrine and Metabolic Diseases, Jinan, Shandong, China
- Shandong Engineering Laboratory of Prevention and Control for Endocrine and Metabolic Diseases, Jinan, Shandong, China
- Department of Endocrinology, Shandong Provincial Hospital, Shandong University, Jinan, Shandong, China
| | - Guodong Liu
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education; Department of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
- Shandong Key Laboratory of Endocrine Metabolism and Aging, Jinan, Shandong, China
- Shandong Institute of Endocrine and Metabolic Diseases, Jinan, Shandong, China
- Shandong Engineering Laboratory of Prevention and Control for Endocrine and Metabolic Diseases, Jinan, Shandong, China
| | - Haiqing Zhang
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education; Department of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
- Shandong Key Laboratory of Endocrine Metabolism and Aging, Jinan, Shandong, China
- Shandong Institute of Endocrine and Metabolic Diseases, Jinan, Shandong, China
- Shandong Engineering Laboratory of Prevention and Control for Endocrine and Metabolic Diseases, Jinan, Shandong, China
| | - Xiude Fan
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education; Department of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
- Shandong Key Laboratory of Endocrine Metabolism and Aging, Jinan, Shandong, China
- Shandong Institute of Endocrine and Metabolic Diseases, Jinan, Shandong, China
- Shandong Engineering Laboratory of Prevention and Control for Endocrine and Metabolic Diseases, Jinan, Shandong, China
| | - Jiajun Zhao
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education; Department of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China.
- Shandong Key Laboratory of Endocrine Metabolism and Aging, Jinan, Shandong, China.
- Shandong Institute of Endocrine and Metabolic Diseases, Jinan, Shandong, China.
- Shandong Engineering Laboratory of Prevention and Control for Endocrine and Metabolic Diseases, Jinan, Shandong, China.
| |
Collapse
|
13
|
Doherty S, Hernandez S, Rikhi R, Mirzai S, De Los Reyes C, McIntosh S, Block RC, Shapiro MD. Lipoprotein(a) as a Causal Risk Factor for Cardiovascular Disease. CURRENT CARDIOVASCULAR RISK REPORTS 2025; 19:8. [PMID: 39980866 PMCID: PMC11836235 DOI: 10.1007/s12170-025-00760-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/04/2025] [Indexed: 02/22/2025]
Abstract
Purpose of Review Lipoprotein(a) [Lp(a)], an atherogenic low-density lipoprotein cholesterol (LDL-C)-like molecule, has emerged as an important risk factor for the development of atherosclerotic cardiovascular disease (ASCVD). This review summarizes the evidence supporting Lp(a) as a causal risk factor for ASCVD and calcific aortic valve stenosis (CAVS). Recent Findings Lp(a) is largely (~ 90%) genetically determined and approximately 20% of the global population has elevated Lp(a). The unique structure of Lp(a) leads to proatherogenic, proinflammatory, and antifibrinolytic properties. Data from epidemiological, genome-wide association, Mendelian randomization, and meta-analyses have shown a clear association between Lp(a) and ASCVD, as well as CAVS. There are emerging data on the association between Lp(a) and ischemic stroke, peripheral arterial disease, and heart failure; however, the associations are not as strong. Summary Several lines of evidence support Lp(a) as a causal risk factor for ASCVD and CAVS. The 2024 National Lipid Association guidelines, 2022 European Atherosclerosis Society, and 2021 Canadian Cardiology Society guidelines recommend testing Lp(a) once in all adults to guide primary prevention efforts. Further studies on cardiovascular outcomes with Lp(a) targeted therapies will provide more insight on causal relationship between Lp(a) and cardiovascular disease.
Collapse
Affiliation(s)
- Sean Doherty
- Department of Internal Medicine, Wake Forest University School of Medicine, Medical Center Boulevard, Winston-Salem, NC 27157 USA
| | - Sebastian Hernandez
- Department of Internal Medicine, Wake Forest University School of Medicine, Medical Center Boulevard, Winston-Salem, NC 27157 USA
| | - Rishi Rikhi
- Center for Prevention of Cardiovascular Disease, Section on Cardiovascular Medicine, Department of Internal Medicine, Medical Center Boulevard, Wake Forest University School of Medicine, Medical Center Boulevard, Winston-Salem, NC 27157 USA
| | - Saeid Mirzai
- Center for Prevention of Cardiovascular Disease, Section on Cardiovascular Medicine, Department of Internal Medicine, Medical Center Boulevard, Wake Forest University School of Medicine, Medical Center Boulevard, Winston-Salem, NC 27157 USA
| | - Chris De Los Reyes
- Department of Public Health Sciences, University of Rochester School of Medicine and Dentistry, 14642 Rochester, NY USA
| | - Scott McIntosh
- Department of Public Health Sciences, Division of Public Health Sciences, University of Rochester Medical Center, Rochester, NY 14642 USA
| | - Robert C. Block
- Department of Public Health Sciences, Division of Epidemiology, Department of Medicine’s Cardiology Division, University of Rochester School of Medicine and Dentistry, Rochester, NY 14642 USA
| | - Michael D. Shapiro
- Center for Prevention of Cardiovascular Disease, Section on Cardiovascular Medicine, Department of Internal Medicine, Medical Center Boulevard, Wake Forest University School of Medicine, Medical Center Boulevard, Winston-Salem, NC 27157 USA
| |
Collapse
|
14
|
Steinmeyer J. Phospholipids and Sphingolipids in Osteoarthritis. Biomolecules 2025; 15:250. [PMID: 40001553 PMCID: PMC11853253 DOI: 10.3390/biom15020250] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2025] [Revised: 02/04/2025] [Accepted: 02/05/2025] [Indexed: 02/27/2025] Open
Abstract
Many studies now emphasize the intricate relationship between lipid metabolism and osteoarthritis (OA), a leading cause of disability. This narrative review examines alterations in the levels of phospholipids (PLs) and sphingolipids (SLs) in synovial fluid (SF), plasma, serum, and articular tissues; discusses their role in joint lubrication, inflammation, and cartilage degradation; and describes their potential as diagnostic markers and therapeutic targets. Key findings include stage-dependent elevated levels of specific PLs and SLs in the SF, blood, and tissue of OA patients, implicating them as possible biomarkers of disease severity and progression. Studies suggest that beyond the involvement of these lipids in joint lubrication, individual species, such as lysophosphatidylcholine (LPC) 16:0, lysophosphatidic acid (LPA), ceramide-1-phosphate (C1P), and sphingosine-1-phosphate (S1P), contribute to pain, inflammation, and degradation of joints through various signaling pathways. Cross-species comparisons suggest that dogs and mice experience similar lipidomic changes during OA as humans, rendering them valuable models for studying lipid-related mechanisms. PLs and SLs in SF appear to originate primarily from the synovial blood capillaries through diffusion. In addition, lipids that are produced locally by fibroblast-like synoviocytes (FLSs) are influenced by cytokines and growth factors that regulate the biosynthesis of PLs for joint lubrication. Emerging research has identified genes such as UGCG and ESYT1 as regulators of lipid metabolism in OA. Further, we examine the suitability of lipids as biomarkers of OA and the potential of targeting the PL and SL pathways to treat OA, emphasizing the need for further research to translate these findings into clinical applications.
Collapse
Affiliation(s)
- Juergen Steinmeyer
- Laboratory for Experimental Orthopaedics, Department of Orthopaedics and Orthopaedic Surgery, Justus Liebig University, 35392 Giessen, Germany
| |
Collapse
|
15
|
Jokesch P, Oskolkova O, Fedorova M, Gesslbauer B, Bochkov V. Contribution of individual phospholipase A 2 enzymes to the cleavage of oxidized phospholipids in human blood plasma. J Lipid Res 2025; 66:100742. [PMID: 39778770 PMCID: PMC11841071 DOI: 10.1016/j.jlr.2025.100742] [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: 08/29/2024] [Revised: 12/16/2024] [Accepted: 01/03/2025] [Indexed: 01/11/2025] Open
Abstract
Phospholipids containing oxidized esterified PUFA residues (OxPLs) are increasingly recognized for multiple biological activities and causative involvement in disease pathogenesis. Pharmacokinetics of these compounds in blood plasma is essentially not studied. Human plasma contains both genuine phospholipases A2 [platelet activating factor acetyl hydrolase (PAF-AH) (also called Lp-PLA2) and secretory phospholipase A2] and multifunctional enzymes capable of removing sn-2 residues in native and oxidized PLs (lecithin-cholesterol acyltransferase, peroxiredoxin-6). The goal of this study was to compare relative activities of different PLA2 enzymes by analyzing cleavage of oxidized 1-palmitoyl-2-arachidonoyl-sn-glycero-phosphatidylcholine (OxPAPC) and oxidized 1-palmitoyl-2-arachidonoyl-sn-glycero-phosphatidylethanolamine (OxPAPE) by diluted plasma in the presence of enzyme inhibitors. We have found that human plasma demonstrated high total PLA2 activity against oxidized PCs and PEs. PAF-AH/Lp-PLA2 played a dominant role in LysoPC and LysoPE production as compared to other enzymes. Molecular species of oxidized 1-palmitoyl-2-arachidonoyl-sn-glycero-phosphatidylcholine and oxidized 1-palmitoyl-2-arachidonoyl-sn-glycero-phosphatidylethanolamine could be divided into three groups according to their degradation rate and sensitivity to PAF-AH/Lp-PLA2 inhibitor darapladib. Oxidatively truncated species were most rapidly metabolized in the presence of plasma; this process was strongly inhibited by darapladib. The rate of degradation of full-length OxPLs depended on the degree of oxygenation. Species containing 1 to 3 oxygen atoms were relatively stable to degradation in plasma, while OxPLs containing > 3 extra oxygens were degraded but at significantly slower rate than truncated species. In contrast to truncated species, degradation of full-length OxPLs with > 3 extra oxygens were only minimally inhibited by darapladib. These data provide further insights into the mechanisms regulating circulating levels of OxPLs and lipid mediators generated by PLA2 cleavage of OxPLs, namely oxylipins and LysoPC.
Collapse
Affiliation(s)
- Philipp Jokesch
- Institute of Pharmaceutical Sciences, Department of Pharmaceutical Chemistry, University of Graz, Graz, Austria
| | - Olga Oskolkova
- Institute of Pharmaceutical Sciences, Department of Pharmaceutical Chemistry, University of Graz, Graz, Austria
| | - Maria Fedorova
- Center of Membrane Biochemistry and Lipid Research, University Hospital Carl Gustav Carus and Faculty of Medicine of TU Dresden, Dresden, Germany
| | - Bernd Gesslbauer
- Institute of Pharmaceutical Sciences, Department of Pharmaceutical Chemistry, University of Graz, Graz, Austria.
| | - Valery Bochkov
- Institute of Pharmaceutical Sciences, Department of Pharmaceutical Chemistry, University of Graz, Graz, Austria; Field of Excellence BioHealth - University of Graz, Graz, Austria.
| |
Collapse
|
16
|
Chen J, Pei B, Shi S. Association between egg consumption and risk of obesity: A comprehensive review: EGG CONSUMPTION AND OBESITY. Poult Sci 2025; 104:104660. [PMID: 39721264 PMCID: PMC11731440 DOI: 10.1016/j.psj.2024.104660] [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: 09/20/2024] [Revised: 12/04/2024] [Accepted: 12/10/2024] [Indexed: 12/28/2024] Open
Abstract
Eggs serve as a vital source of high-quality protein and lipids in the human diet, contributing significantly to nutritional intake; however, the relation between egg intake and health risks has been controversial. This study aimed to assess the relationship between egg intake and obesity and the effects of the various nutrients in eggs on obesity were separately investigated. This review involved searching Scopus, PubMed, Google Scholar for relevant articles from 2002 to 2022. Studies suggested that moderate egg consumption exerts little effect on blood lipid levels, that due to the body regulates endogenous cholesterol production in response to the external cholesterol intake. Furthermore, certain studies also verified that the presence of other nutrients in eggs, such as lecithin, unsaturated fatty acids, and apolipoproteins, not only does not contribute to elevated blood lipids but also plays a role in regulating lipid metabolism to prevent obesity. Additionally, the study reveals that different cooking methods significantly impact the nutritional composition of eggs, with soft-boiled eggs generally being the most advantageous for human health. This article reveals that dietary cholesterol or moderate egg intake was not significantly associated with a higher risk of obesity in healthy adults. Nevertheless, cholesterol-sensitive individuals should ensure moderate cholesterol intake.
Collapse
Affiliation(s)
- Jinglong Chen
- Jiangsu Institute of Poultry Science, Yangzhou, 225125, China.
| | - Bixuan Pei
- Jiangsu Institute of Poultry Science, Yangzhou, 225125, China.
| | - Shourong Shi
- Jiangsu Institute of Poultry Science, Yangzhou, 225125, China.
| |
Collapse
|
17
|
Ramírez-Melo LM, Estrada-Luna D, Rubio-Ruiz ME, Castañeda-Ovando A, Fernández-Martínez E, Jiménez-Osorio AS, Pérez-Méndez Ó, Carreón-Torres E. Relevance of Lipoprotein Composition in Endothelial Dysfunction and the Development of Hypertension. Int J Mol Sci 2025; 26:1125. [PMID: 39940892 PMCID: PMC11817739 DOI: 10.3390/ijms26031125] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2024] [Revised: 01/18/2025] [Accepted: 01/21/2025] [Indexed: 02/16/2025] Open
Abstract
Endothelial dysfunction and chronic inflammation are determining factors in the development and progression of chronic degenerative diseases, such as hypertension and atherosclerosis. Among the shared pathophysiological characteristics of these two diseases is a metabolic disorder of lipids and lipoproteins. Therefore, the contents and quality of the lipids and proteins of lipoproteins become the targets of therapeutic objective. One of the stages of lipoprotein formation occurs through the incorporation of dietary lipids by enterocytes into the chylomicrons. Consequently, the composition, structure, and especially the properties of lipoproteins could be modified through the intake of bioactive compounds. The objective of this review is to describe the roles of the different lipid and protein components of lipoproteins and their receptors in endothelial dysfunction and the development of hypertension. In addition, we review the use of some non-pharmacological treatments that could improve endothelial function and/or prevent endothelial damage. The reviewed information contributes to the understanding of lipoproteins as vehicles of regulatory factors involved in the modulation of inflammatory and hemostatic processes, the attenuation of oxidative stress, and the neutralization of toxins, rather than only cholesterol and phospholipid transporters. For this review, a bibliographic search was carried out in different online metabases.
Collapse
Affiliation(s)
- Lisette Monsibaez Ramírez-Melo
- Nutrition Academic Area Instituto de Ciencias de la Salud, Universidad Autónoma del Estado de Hidalgo, Circuito Ex Hacienda La Concepción S/N, Carretera Pachuca-Actopan, San Agustín Tlaxiaca 42160, Hidalgo, Mexico;
| | - Diego Estrada-Luna
- Nursing Academic Area, Instituto de Ciencias de la Salud, Universidad Autónoma del Estado de Hidalgo, Circuito Ex Hacienda La Concepción S/N, Carretera Pachuca-Actopan, San Agustín Tlaxiaca 42160, Hidalgo, Mexico; (D.E.-L.); (A.S.J.-O.)
| | - María Esther Rubio-Ruiz
- Department of Physiology, Instituto Nacional de Cardiología Ignacio Chávez, Juan Badiano 1, Tlalpan, Mexico City 14080, Mexico;
| | - Araceli Castañeda-Ovando
- Chemistry Academic Area, Instituto de Ciencias Básicas e Ingeniería, Universidad Autónoma del Estado de Hidalgo, Pachuca 42039, Hidalgo, Mexico;
| | - Eduardo Fernández-Martínez
- Medicine Academic Area, Instituto de Ciencias de la Salud, Universidad Autónoma del Estado de Hidalgo, Pachuca 42039, Hidalgo, Mexico;
| | - Angélica Saraí Jiménez-Osorio
- Nursing Academic Area, Instituto de Ciencias de la Salud, Universidad Autónoma del Estado de Hidalgo, Circuito Ex Hacienda La Concepción S/N, Carretera Pachuca-Actopan, San Agustín Tlaxiaca 42160, Hidalgo, Mexico; (D.E.-L.); (A.S.J.-O.)
| | - Óscar Pérez-Méndez
- Department of Molecular Biology, Instituto Nacional de Cardiología Ignacio Chávez, Juan Badiano 1, Mexico City 14080, Mexico;
- Tecnológico de Monterrey, Campus Ciudad de México, Mexico City 14380, Mexico
| | - Elizabeth Carreón-Torres
- Department of Molecular Biology, Instituto Nacional de Cardiología Ignacio Chávez, Juan Badiano 1, Mexico City 14080, Mexico;
| |
Collapse
|
18
|
Bhatia HS, Wandel S, Willeit P, Lesogor A, Bailey K, Ridker PM, Nestel P, Simes J, Tonkin A, Schwartz GG, Colhoun H, Wanner C, Tsimikas S. Independence of Lipoprotein(a) and Low-Density Lipoprotein Cholesterol-Mediated Cardiovascular Risk: A Participant-Level Meta-Analysis. Circulation 2025; 151:312-321. [PMID: 39492722 PMCID: PMC11771346 DOI: 10.1161/circulationaha.124.069556] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Accepted: 09/24/2024] [Indexed: 11/05/2024]
Abstract
BACKGROUND Low-density lipoprotein cholesterol (LDL-C) and lipoprotein(a) (Lp[a]) levels are independently associated with atherosclerotic cardiovascular disease (ASCVD). However, the relationship between Lp(a) level, LDL-C level, and ASCVD risk at different thresholds is not well defined. METHODS A participant-level meta-analysis of 27 658 participants enrolled in 6 placebo-controlled statin trials was performed to assess the association of LDL-C and Lp(a) levels with risk of fatal or nonfatal coronary heart disease events, stroke, or any coronary or carotid revascularization (ASCVD). The multivariable-adjusted association between baseline Lp(a) level and ASCVD risk was modeled continuously using generalized additive models, and the association between baseline LDL-C level and ASCVD risk by baseline Lp(a) level by Cox proportional hazards models with random effects. The joint association between Lp(a) level and statin-achieved LDL-C level with ASCVD risk was evaluated using Cox proportional hazards models. RESULTS Compared with an Lp(a) level of 5 mg/dL, increasing levels of Lp(a) were log-linearly associated with ASCVD risk in statin- and placebo-treated patients. Among statin-treated individuals, those with Lp(a) level >50 mg/dL (≈125 nmol/L) had increased risk across all quartiles of achieved LDL-C level and absolute change in LDL-C level. Even among those with the lowest quartile of achieved LDL-C level (3.1-77.0 mg/dL), those with Lp(a) level >50 mg/dL had greater ASCVD risk (hazard ratio, 1.38 [95% CI, 1.06-1.79]) than those with Lp(a) level ≤50 mg/dL. The greatest risk was observed with both Lp(a) level >50 mg/dL and LDL-C level in the fourth quartile (hazard ratio, 1.90 [95% CI, 1.46-2.48]). CONCLUSIONS These findings demonstrate the independent and additive nature of Lp(a) and LDL-C levels for ASCVD risk, and that LDL-C lowering does not fully offset Lp(a)-mediated risk.
Collapse
Affiliation(s)
- Harpreet S. Bhatia
- Division of Cardiology, Department of Medicine, University of California, San Diego, La Jolla (H.S.B., S.T.)
| | - Simon Wandel
- Novartis Pharma AG, Basel, Switzerland (S.W., A.L., K.B.)
| | - Peter Willeit
- Department of Medical Statistics, Informatics and Health Economics at the Medical University of Innsbruck, Austria (P.W.)
| | | | - Keith Bailey
- Novartis Pharma AG, Basel, Switzerland (S.W., A.L., K.B.)
| | - Paul M. Ridker
- Brigham and Women’s Hospital, Harvard Medical School, Boston, MA (P.M.R.)
| | - Paul Nestel
- Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia (P.N.)
| | - John Simes
- NHMRC Clinical Trials Centre, University of Sydney, NSW, Australia (J.S.)
| | - Andrew Tonkin
- School of Public Health and Preventive Medicine, Monash University, Melbourne, Victoria, Australia (A.T.)
| | - Gregory G. Schwartz
- Division of Cardiology, Rocky Mountain Regional VA Medical Center and University of Colorado School of Medicine, Aurora (G.G.S.)
| | - Helen Colhoun
- MRC Human Genetics Unit, Centre for Genomic and Experimental Medicine, MRC Institute of Genetics & Molecular Medicine, Edinburgh, UK (H.C.)
| | - Christoph Wanner
- Division of Nephrology, Department of Internal Medicine and Comprehensive Heart Failure Centre, University Hospital of Würzburg, Germany (C.W.)
| | - Sotirios Tsimikas
- Division of Cardiology, Department of Medicine, University of California, San Diego, La Jolla (H.S.B., S.T.)
| |
Collapse
|
19
|
Choleva M, Antonopoulou S, Fragopoulou E. Winery By-Products In Vitro and In Vivo Effects on Atherothrombotic Markers: Focus on Platelet-Activating Factor. FRONT BIOSCI-LANDMRK 2025; 30:25859. [PMID: 39862073 DOI: 10.31083/fbl25859] [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: 05/30/2024] [Revised: 08/21/2024] [Accepted: 08/28/2024] [Indexed: 01/27/2025]
Abstract
Platelet aggregation and inflammation play a crucial role in atherothrombosis. Wine contains micro-constituents of proper quality and quantity that exert cardioprotective actions, partly through inhibiting platelet-activating factor (PAF), a potent inflammatory and thrombotic lipid mediator. However, wine cannot be consumed extensively due to the presence of ethanol. Alternatively, winery by-products are abundant in similar-to-wine micro-constituents that could be used in food fortification and dietary supplements. Also, the vinification process produces millions of tons of by-products worldwide, posing an environmental matter of waste management. Therefore, the purpose of this literature review is to update the existing data concerning the in vitro anti-platelet and anti-inflammatory properties of winery by-product extracts and their possible health effects through controlled clinical trials in humans, specifically focused on their effects on PAF's actions. Data from in vitro studies report that winery by-product compounds are able to inhibit platelet aggregation against several aggregation factors, as well as to downregulate inflammatory markers. Among their actions, extracts or phenolic compounds present in winery by-products inhibit PAF's actions, a potent inflammatory and thrombotic mediator. Similar conclusions have been drawn from human supplementation studies, which suggest that winery by-product extracts may have beneficial biological effects on the cardiovascular system. Evidence from long-term studies shows that consumption may lower total and low density lipoprotein (LDL) cholesterol, improve insulin sensitivity, decrease lipid and protein oxidative damage, enhance antioxidant capacity, and have mild anti-inflammatory action toward reducing cytokine expression and levels. Data from the limited postprandial studies report that the acute consumption of winery by-product extracts improves glycemic response and reduces platelet reactivity to aggregatory stimuli. Although wine extracts and phenolic compounds have been reported to inhibit PAF's actions and reduce the activity of its biosynthetic enzymes, no data exist concerning the influence of winery by-product extracts. In the future, additional long-term randomized controlled trials or postprandial studies are needed to draw definitive conclusions and establish a viable cardioprotective strategy that incorporates the sustainable use of winery by-products.
Collapse
Affiliation(s)
- Maria Choleva
- Department of Nutrition and Dietetics, School of Health Sciences and Education, Harokopio University, 17676 Athens, Greece
| | - Smaragdi Antonopoulou
- Department of Nutrition and Dietetics, School of Health Sciences and Education, Harokopio University, 17676 Athens, Greece
| | - Elizabeth Fragopoulou
- Department of Nutrition and Dietetics, School of Health Sciences and Education, Harokopio University, 17676 Athens, Greece
| |
Collapse
|
20
|
Palmieri M, Maraka S, Spencer HJ, Thostenson JD, Dishongh K, Knox M, Ussery B, Byrd J, Kuipers JK, Abedzadeh-Anaraki S, Duvoor C, Mao Y, Menon L, Williams JS, Manolagas SC, Jilka RL, Ambrogini E. Plasma levels of anti phosphocholine IgM antibodies are negatively correlated with bone mineral density in humans. Sci Rep 2025; 15:2109. [PMID: 39814831 PMCID: PMC11735633 DOI: 10.1038/s41598-025-85624-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Accepted: 01/06/2025] [Indexed: 01/18/2025] Open
Abstract
Phosphatidylcholine is a ubiquitous phospholipid. It contains a phosphocholine (PC) headgroup and polyunsaturated fatty acids that, when oxidized, form reactive oxidized phospholipids (PC-OxPLs). PC-OxPLs are pathogenic in multiple diseases and neutralized by anti-PC IgM antibodies. The levels of anti-PC IgM increase as the levels of PC-OxPLs increase and, in humans, are inversely correlated with the incidence of cardiovascular diseases and steatohepatitis. PC-OxPLs also decrease bone mass in mice. Overexpression of anti-PC IgM ameliorates atherosclerosis and steatohepatitis, increases bone mass in young mice, and protects against high fat diet- and age-associated osteoporosis. We investigated the relationship between anti-PC IgM plasma levels and bone mineral density (BMD) in a cross-sectional study of 247 participants [mean age: 65.5 (± 8.6) years] without medical conditions known to influence BMD or antibody production. Anti-PC IgM levels negatively correlated with both T- and Z-scores at the lumbar spine, femur and, to a lesser extent, the forearm. These correlations were maintained after adjustment for age, race, and sex. These results raise the possibility that higher levels of anti-PC IgM in patients with lower BMD reflect exposure to higher levels of PC-OxPLs, which are known to affect bone mass, and could be a novel risk marker for osteoporosis.
Collapse
Affiliation(s)
- Michela Palmieri
- Division of Endocrinology and Metabolism and Center for Musculoskeletal Disease Research, University of Arkansas for Medical Sciences, 4301 W. Markham, #587, Little Rock, AR, 72205, USA
| | - Spyridoula Maraka
- Division of Endocrinology and Metabolism and Center for Musculoskeletal Disease Research, University of Arkansas for Medical Sciences, 4301 W. Markham, #587, Little Rock, AR, 72205, USA
- Central Arkansas Veterans Healthcare System, Little Rock, AR, USA
| | - Horace J Spencer
- Department of Biostatistics, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Jeff D Thostenson
- Department of Biostatistics, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | | | - Micheal Knox
- Central Arkansas Veterans Healthcare System, Little Rock, AR, USA
| | - Betty Ussery
- Central Arkansas Veterans Healthcare System, Little Rock, AR, USA
| | - Jesse Byrd
- Central Arkansas Veterans Healthcare System, Little Rock, AR, USA
| | | | | | | | - Yuanjie Mao
- Diabetes Institute, Ohio University, Athens, OH, USA
| | - Lakshmi Menon
- Division of Endocrinology and Metabolism and Center for Musculoskeletal Disease Research, University of Arkansas for Medical Sciences, 4301 W. Markham, #587, Little Rock, AR, 72205, USA
| | - James S Williams
- Central Arkansas Veterans Healthcare System, Little Rock, AR, USA
| | - Stavros C Manolagas
- Division of Endocrinology and Metabolism and Center for Musculoskeletal Disease Research, University of Arkansas for Medical Sciences, 4301 W. Markham, #587, Little Rock, AR, 72205, USA
| | - Robert L Jilka
- Division of Endocrinology and Metabolism and Center for Musculoskeletal Disease Research, University of Arkansas for Medical Sciences, 4301 W. Markham, #587, Little Rock, AR, 72205, USA
| | - Elena Ambrogini
- Division of Endocrinology and Metabolism and Center for Musculoskeletal Disease Research, University of Arkansas for Medical Sciences, 4301 W. Markham, #587, Little Rock, AR, 72205, USA.
- Central Arkansas Veterans Healthcare System, Little Rock, AR, USA.
| |
Collapse
|
21
|
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
|
22
|
Gallo A, Le Goff W, Santos RD, Fichtner I, Carugo S, Corsini A, Sirtori C, Ruscica M. Hypercholesterolemia and inflammation-Cooperative cardiovascular risk factors. Eur J Clin Invest 2025; 55:e14326. [PMID: 39370572 PMCID: PMC11628670 DOI: 10.1111/eci.14326] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Accepted: 09/02/2024] [Indexed: 10/08/2024]
Abstract
BACKGROUND Maintaining low concentrations of plasma low-density lipoprotein cholesterol (LDLc) over time decreases the number of LDL particles trapped within the artery wall, slows the progression of atherosclerosis and delays the age at which mature atherosclerotic plaques develop. This substantially reduces the lifetime risk of atherosclerotic cardiovascular disease (ASCVD) events. In this context, plaque development and vulnerability result not only from lipid accumulation but also from inflammation. RESULTS Changes in the composition of immune cells, including macrophages, dendritic cells, T cells, B cells, mast cells and neutrophils, along with altered cytokine and chemokine release, disrupt the equilibrium between inflammation and anti-inflammatory mechanisms at plaque sites. Considering that it is not a competition between LDLc and inflammation, but instead that they are partners in crime, the present narrative review aims to give an overview of the main inflammatory molecular pathways linked to raised LDLc concentrations and to describe the impact of lipid-lowering approaches on the inflammatory and lipid burden. Although remarkable changes in LDLc are driven by the most recent lipid lowering combinations, the relative reduction in plasma C-reactive protein appears to be independent of the magnitude of LDLc lowering. CONCLUSION Identifying clinical biomarkers of inflammation (e.g. interleukin-6) and possible targets for therapy holds promise for monitoring and reducing the ASCVD burden in suitable patients.
Collapse
Affiliation(s)
- Antonio Gallo
- Lipidology and Cardiovascular Prevention Unit, Department of Nutrition, APHP, Hôpital Pitié‐SalpètriêreSorbonne Université, INSERM UMR1166ParisFrance
| | - Wilfried Le Goff
- Lipidology and Cardiovascular Prevention Unit, Department of Nutrition, APHP, Hôpital Pitié‐SalpètriêreSorbonne Université, INSERM UMR1166ParisFrance
| | - Raul D. Santos
- Academic Research Organization Hospital Israelita Albert Einstein and Lipid Clinic Heart Institute (InCor)University of Sao Paulo Medical School HospitalSao PauloBrazil
| | - Isabella Fichtner
- Department of Pharmacological and Biomolecular Sciences “Rodolfo Paoletti”Università degli Studi di MilanoMilanItaly
| | - Stefano Carugo
- Department of Cardio‐Thoracic‐Vascular DiseasesFoundation IRCCS Cà Granda Ospedale Maggiore PoliclinicoMilanItaly
- Department of Clinical Sciences and Community HealthUniversità degli Studi di MilanoMilanItaly
| | - Alberto Corsini
- Department of Pharmacological and Biomolecular Sciences “Rodolfo Paoletti”Università degli Studi di MilanoMilanItaly
| | - Cesare Sirtori
- Department of Pharmacological and Biomolecular Sciences “Rodolfo Paoletti”Università degli Studi di MilanoMilanItaly
| | - Massimiliano Ruscica
- Department of Pharmacological and Biomolecular Sciences “Rodolfo Paoletti”Università degli Studi di MilanoMilanItaly
- Department of Cardio‐Thoracic‐Vascular DiseasesFoundation IRCCS Cà Granda Ospedale Maggiore PoliclinicoMilanItaly
| |
Collapse
|
23
|
Bhatia HS, Dweck MR, Craig N, Capoulade R, Pibarot P, Trainor PJ, Whelton SP, Rikhi R, Lidani KCF, Post WS, Tsai MY, Criqui MH, Shapiro MD, Budoff MJ, DeFilippis AP, Thanassoulis G, Tsimikas S. Oxidized Phospholipids and Calcific Aortic Valvular Disease. J Am Coll Cardiol 2024; 84:2430-2441. [PMID: 39545902 DOI: 10.1016/j.jacc.2024.08.070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 08/12/2024] [Accepted: 08/15/2024] [Indexed: 11/17/2024]
Abstract
BACKGROUND Oxidized phospholipids (OxPLs) are carried by apolipoprotein B-100-containing lipoproteins (OxPL-apoB) including lipoprotein(a) (Lp[a]). Both OxPL-apoB and Lp(a) have been associated with calcific aortic valve disease (CAVD). OBJECTIVES This study aimed to evaluate the associations between OxPL-apoB, Lp(a) and the prevalence, incidence, and progression of CAVD. METHODS OxPL-apoB and Lp(a) were evaluated in MESA (Multi-Ethnic Study of Atherosclerosis) and a participant-level meta-analysis of 4 randomized trials of participants with established aortic stenosis (AS). In MESA, the association of OxPL-apoB and Lp(a) with aortic valve calcium (AVC) at baseline and 9.5 years was evaluated using multivariable ordinal regression models. In the meta-analysis, the association between OxPL-apoB and Lp(a) with AS progression (annualized change in peak aortic valve jet velocity) was evaluated using multivariable linear regression models. RESULTS In MESA, both OxPL-apoB and Lp(a) were associated with prevalent AVC (OR per SD: 1.19 [95% CI: 1.07-1.32] and 1.13 [95% CI: 1.01-1.27], respectively) with a significant interaction between the two (P < 0.01). Both OxPL-apoB and Lp(a) were associated with incident AVC at 9.5 years when evaluated individually (interaction P < 0.01). The OxPL-apoB∗Lp(a) interaction demonstrated higher odds of prevalent and incident AVC for OxPL-apoB with increasing Lp(a) levels. In the meta-analysis, when analyzed separately, both OxPL-apoB and Lp(a) were associated with faster increase in peak aortic valve jet velocity, but when evaluated together, only OxPL-apoB remained significant (ß: 0.07; 95% CI: 0.01-0.12). CONCLUSIONS OxPL-apoB is a predictor of the presence, incidence, and progression of AVC and established AS, particularly in the setting of elevated Lp(a) levels, and may represent a novel therapeutic target for CAVD.
Collapse
Affiliation(s)
- Harpreet S Bhatia
- Division of Cardiology, Department of Medicine, University of California-San Diego, La Jolla, California, USA
| | - Marc R Dweck
- Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, United Kingdom
| | - Neil Craig
- Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, United Kingdom
| | - Romain Capoulade
- Nantes Université, CHU Nantes, CNRS, INSERM, l'institut du thorax, Nantes, France
| | - Philippe Pibarot
- Department of Cardiology, Institut universitaire de cardiologie et de pneumologie de Québec, Laval University, Québec, Québec, Canada
| | - Patrick J Trainor
- Department of Chemistry and Biochemistry, New Mexico State University, Las Cruces, New Mexico, USA
| | - Seamus P Whelton
- Division of Cardiology, Department of Medicine, Johns Hopkins University, Baltimore, Maryland, USA
| | - Rishi Rikhi
- Section of Cardiovascular Medicine, Department of Internal Medicine, Wake Forest University School of Medicine, Winston Salem, North Carolina, USA
| | - Karita C F Lidani
- Division of Cardiovascular Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Wendy S Post
- Division of Cardiology, Department of Medicine, Johns Hopkins University, Baltimore, Maryland, USA
| | - Michael Y Tsai
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, Minnesota, USA
| | - Michael H Criqui
- Division of Cardiology, Department of Medicine, University of California-San Diego, La Jolla, California, USA; Division of Preventive Medicine, Department of Family Medicine, University of California-San Diego, La Jolla, California, USA
| | - Michael D Shapiro
- Section of Cardiovascular Medicine, Department of Internal Medicine, Wake Forest University School of Medicine, Winston Salem, North Carolina, USA
| | - Matthew J Budoff
- Division of Cardiology, Lundquist Institute at Harbor-UCLA Medical Center, Torrance, California, USA
| | - Andrew P DeFilippis
- Division of Cardiovascular Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - George Thanassoulis
- Department of Medicine, Division of Experimental Medicine, McGill University Health Center, Montreal, Québec, Canada
| | - Sotirios Tsimikas
- Division of Cardiology, Department of Medicine, University of California-San Diego, La Jolla, California, USA.
| |
Collapse
|
24
|
Strandberg TE, Kovanen PT, Lloyd-Jones DM, Raal FJ, Santos RD, Watts GF. Drugs for dyslipidaemia: the legacy effect of the Scandinavian Simvastatin Survival Study (4S). Lancet 2024; 404:2462-2475. [PMID: 39577453 DOI: 10.1016/s0140-6736(24)02089-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Revised: 09/16/2024] [Accepted: 09/19/2024] [Indexed: 11/24/2024]
Abstract
Since the discovery of statins and the Scandinavian Simvastatin Survival Study (4S) results three decades ago, remarkable advances have been made in the treatment of dyslipidaemia, a major risk factor for atherosclerotic cardiovascular disease. Safe and effective statins remain the cornerstone of therapeutic approach for this indication, including for children with genetic dyslipidaemia, and are one of the most widely prescribed drugs in the world. However, despite the affordability of generic statins, they remain underutilised worldwide. The use of ezetimibe to further decrease plasma LDL cholesterol and the targeting of other atherogenic lipoproteins, such as triglyceride-rich lipoproteins and lipoprotein(a), are likely to be required to further reduce atherosclerotic cardiovascular disease events. Drugs directed at these lipoproteins, including gene silencing and editing methods that durably suppress the production of proteins, such as PCSK9 and ANGPTL3, open novel therapeutic options to further reduce the development of atherosclerotic cardiovascular disease.
Collapse
Affiliation(s)
- Timo E Strandberg
- University of Helsinki and Helsinki University Hospital, Helsinki, Finland; University of Oulu, Center for Life Course Health Research, Oulu, Finland.
| | | | - Donald M Lloyd-Jones
- Department of Preventive Medicine and Department of Medicine (Cardiology), Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Frederick J Raal
- Division of Endocrinology and Metabolism, University of the Witwatersrand, Johannesburg, South Africa
| | - Raul D Santos
- Academic Research Organization, Hospital Israelita Albert Einstein, São Paulo, Brazil; Lipid Clinic Heart Institute (InCor) University of São Paulo Medical School Hospital, São Paulo, Brazil
| | - Gerald F Watts
- School of Medicine, University of Western Australia, Perth, WA, Australia; Cardiometabolic Service, Department of Cardiology and Internal Medicine, Royal Perth Hospital, Perth, WA, Australia
| |
Collapse
|
25
|
Vo DK, Trinh KTL. Emerging Biomarkers in Metabolomics: Advancements in Precision Health and Disease Diagnosis. Int J Mol Sci 2024; 25:13190. [PMID: 39684900 DOI: 10.3390/ijms252313190] [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: 11/19/2024] [Revised: 12/01/2024] [Accepted: 12/06/2024] [Indexed: 12/18/2024] Open
Abstract
Metabolomics has come to the fore as an efficient tool in the search for biomarkers that are critical for precision health approaches and improved diagnostics. This review will outline recent advances in biomarker discovery based on metabolomics, focusing on metabolomics biomarkers reported in cancer, neurodegenerative disorders, cardiovascular diseases, and metabolic health. In cancer, metabolomics provides evidence for unique oncometabolites that are important for early disease detection and monitoring of treatment responses. Metabolite profiling for conditions such as neurodegenerative and mental health disorders can offer early diagnosis and mechanisms into the disease especially in Alzheimer's and Parkinson's diseases. In addition to these, lipid biomarkers and other metabolites relating to cardiovascular and metabolic disorders are promising for patient stratification and personalized treatment. The gut microbiome and environmental exposure also feature among the influential factors in biomarker discovery because they sculpt individual metabolic profiles, impacting overall health. Further, we discuss technological advances in metabolomics, current clinical applications, and the challenges faced by metabolomics biomarker validation toward precision medicine. Finally, this review discusses future opportunities regarding the integration of metabolomics into routine healthcare to enable preventive and personalized approaches.
Collapse
Affiliation(s)
- Dang-Khoa Vo
- College of Pharmacy, Gachon University, 191 Hambakmoe-ro, Yeonsu-gu, Incheon 21936, Republic of Korea
| | - Kieu The Loan Trinh
- BioNano Applications Research Center, Gachon University, 1342 Seongnam-daero, Sujeong-gu, Seongnam-si 13120, Gyeonggi-do, Republic of Korea
| |
Collapse
|
26
|
Mokhtari I, Aljutaily T, Aljumayi H, Radhi KS, Almutairi AS, Barakat H, Khalifa I, Amrani S, Harnafi H. Metabolic Effects of Loquat Juice ( Eriobotrya japonica Lindl Mkarkeb Variety) on Lipid Homeostasis, Liver Steatosis, and Oxidative Stress in Hyperlipidemic Mice Fed a High-Fat-High-Fructose Diet. Metabolites 2024; 14:592. [PMID: 39590828 PMCID: PMC11596324 DOI: 10.3390/metabo14110592] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Revised: 10/29/2024] [Accepted: 10/31/2024] [Indexed: 11/28/2024] Open
Abstract
BACKGROUND Loquat fruit is consumed for its flavorful taste and a rich array of health-promoting compounds like phenolics, flavonoids, and carotenoids. This study aimed at the biochemical characterization of fresh juice from the Moroccan Mkarkeb variety of loquat and evaluating its effects on lipid homeostasis and liver steatosis in hyperlipidemic mice. METHODS The biochemical characterization followed AOAC methods. In vivo study involved hyperlipidemic mice fed a high-fat, high-fructose diet for 6 weeks and treated with loquat juice at 3.5 and 7 mL kg-1 or fenofibrate at 4 mg·kg-1. The concentrations of lipids in plasma, liver, adipose tissue, feces, and bile and blood glucose levels were quantified. Liver steatosis was visually examined and confirmed histologically, and liver injury markers (AST, ALT, ALP, LDH, and TB) were measured. Liver oxidative stress was assessed by measuring MDA content and antioxidative enzyme activities. RESULTS Our findings indicate that fresh loquat juice is poor in fat and protein and contains moderate sugars with a low energy value (40.82 ± 0.25 kcal/100 g). It is also rich in minerals, vitamin C, phenolic acids, flavonoids, and carotenoids. The juice effectively restored lipid metabolism by enhancing reverse cholesterol transport and lowering LDL-cholesterol, triglycerides, and the atherogenic index. The studied juice decreases blood glucose and prevents weight gain and lipid accumulation in the liver and adipose tissue. The juice prevents lipotoxicity-induced liver injury, corrects toxicity markers, and improves the liver's morphological and histological structures. It also reduces oxidative stress by lowering MDA and activating SOD and catalase. CONCLUSIONS The juice holds high nutritional and medicinal value, potentially preventing lipid disorders and cardiovascular issues.
Collapse
Affiliation(s)
- Imane Mokhtari
- Laboratory of Bioresources, Biotechnologies, Ethnopharmacology and Health, Faculty of Sciences, University Mohamed I, Oujda 60 000, Morocco; (I.M.); (S.A.); (H.H.)
| | - Thamer Aljutaily
- Department of Food Science and Human Nutrition, College of Agriculture and Food, Qassim University, Buraydah 51452, Saudi Arabia;
| | - Huda Aljumayi
- Department of Food Science and Nutrition, College of Sciences, Taif University, P.O. Box 11099, Taif 21944, Saudi Arabia; (H.A.); (K.S.R.)
| | - Khadija S. Radhi
- Department of Food Science and Nutrition, College of Sciences, Taif University, P.O. Box 11099, Taif 21944, Saudi Arabia; (H.A.); (K.S.R.)
| | - Abdulkarim S. Almutairi
- Al Rass General Hospital, Qassim Health Cluster, Ministry of Health, King Khalid District, Al Rass, Saudi Arabia;
| | - Hassan Barakat
- Department of Food Science and Human Nutrition, College of Agriculture and Food, Qassim University, Buraydah 51452, Saudi Arabia;
| | - Ibrahim Khalifa
- Food Technology Department, Faculty of Agriculture, Benha University, Moshtohor 13736, Egypt;
| | - Souliman Amrani
- Laboratory of Bioresources, Biotechnologies, Ethnopharmacology and Health, Faculty of Sciences, University Mohamed I, Oujda 60 000, Morocco; (I.M.); (S.A.); (H.H.)
| | - Hicham Harnafi
- Laboratory of Bioresources, Biotechnologies, Ethnopharmacology and Health, Faculty of Sciences, University Mohamed I, Oujda 60 000, Morocco; (I.M.); (S.A.); (H.H.)
| |
Collapse
|
27
|
Tsimikas S. Defining the Role of Lp(a) in High-Risk Plaques: The Road to Validation of IVUS, NIRS, OCT, and CTA Approaches. Circ Cardiovasc Imaging 2024; 17:e017613. [PMID: 39561227 DOI: 10.1161/circimaging.124.017613] [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] [Indexed: 11/21/2024]
|
28
|
Björnson E, Adiels M, Borén J, Packard CJ. Lipoprotein(a) is a highly atherogenic lipoprotein: pathophysiological basis and clinical implications. Curr Opin Cardiol 2024; 39:503-510. [PMID: 39360655 DOI: 10.1097/hco.0000000000001170] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/04/2024]
Abstract
PURPOSE OF REVIEW Lipoprotein(a) has been identified as a causal risk factor for atherosclerotic cardiovascular disease (ASCVD) and aortic valve stenosis. However, as reviewed here, there is ongoing debate as to the key pathogenic features of Lp(a) particles and the degree of Lp(a) atherogenicity relative to low-density lipoprotein (LDL). RECENT FINDINGS Genetic analyses have revealed that Lp(a) on a per-particle basis is markedly (about six-fold) more atherogenic than LDL. Oxidized phospholipids carried on Lp(a) have been found to have substantial pro-inflammatory properties triggering pathways that may contribute to atherogenesis. Whether the strength of association of Lp(a) with ASCVD risk is dependent on inflammatory status is a matter of current debate and is critical to implementing intervention strategies. Contradictory reports continue to appear, but most recent studies in large cohorts indicate that the relationship of Lp(a) to risk is independent of C-reactive protein level. SUMMARY Lp(a) is a highly atherogenic lipoprotein and a viable target for intervention in a significant proportion of the general population. Better understanding the basis of its enhanced atherogenicity is important for risk assessment and interpreting intervention trials.
Collapse
Affiliation(s)
| | - Martin Adiels
- Department of Molecular and Clinical Medicine
- School of Public Health and Community Medicine, Institute of Medicine, University of Gothenburg, Gothenburg, Sweden
| | - Jan Borén
- Department of Molecular and Clinical Medicine
| | - Chris J Packard
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, UK
| |
Collapse
|
29
|
Aguayo-Morales H, Poblano J, Berlanga L, Castillo-Tobías I, Silva-Belmares SY, Cobos-Puc LE. Plant Antioxidants: Therapeutic Potential in Cardiovascular Diseases. COMPOUNDS 2024; 4:479-502. [DOI: 10.3390/compounds4030029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
Abstract
Cardiovascular diseases (CVDs) are a global health problem. The mortality associated with them is one of the highest. Essentially, CVDs occur when the heart or blood vessels are damaged. Oxidative stress is an imbalance between the production of reactive oxygen species (free radicals) and antioxidant defenses. Increased production of reactive oxygen species can cause cardiac and vascular injuries, leading to CVDs. Antioxidant therapy has been shown to have beneficial effects on CVDs. Plants are a rich source of bioactive antioxidants on our planet. Several classes of these compounds have been identified. Among them, carotenoids and phenolic compounds are the most potent antioxidants. This review summarizes the role of some carotenoids (a/β-carotene, lycopene and lutein), polyphenols such as phenolic acids (caffeic, p-coumaric, ferulic and chlorogenic acids), flavonoids (quercetin, kaempferol and epigallocatechin gallate), and hydroxytyrosol in mitigating CVDs by studying their biological antioxidant mechanisms. Through detailed analysis, we aim to provide a deeper understanding of how these natural compounds can be integrated into cardiovascular health strategies to help reduce the overall burden of CVD.
Collapse
Affiliation(s)
- Hilda Aguayo-Morales
- Facultad de Ciencias Químicas, Unidad Saltillo, Universidad Autónoma de Coahuila, Boulevard Venustiano Carranza S/N Esquina Con Ing, José Cárdenas Valdés, República Oriente, Saltillo 25290, Mexico
| | - Joan Poblano
- Facultad de Ciencias Químicas, Unidad Saltillo, Universidad Autónoma de Coahuila, Boulevard Venustiano Carranza S/N Esquina Con Ing, José Cárdenas Valdés, República Oriente, Saltillo 25290, Mexico
| | - Lia Berlanga
- Facultad de Ciencias Químicas, Unidad Saltillo, Universidad Autónoma de Coahuila, Boulevard Venustiano Carranza S/N Esquina Con Ing, José Cárdenas Valdés, República Oriente, Saltillo 25290, Mexico
| | - Ileana Castillo-Tobías
- Facultad de Ciencias Químicas, Unidad Saltillo, Universidad Autónoma de Coahuila, Boulevard Venustiano Carranza S/N Esquina Con Ing, José Cárdenas Valdés, República Oriente, Saltillo 25290, Mexico
| | - Sonia Yesenia Silva-Belmares
- Facultad de Ciencias Químicas, Unidad Saltillo, Universidad Autónoma de Coahuila, Boulevard Venustiano Carranza S/N Esquina Con Ing, José Cárdenas Valdés, República Oriente, Saltillo 25290, Mexico
| | - Luis E. Cobos-Puc
- Facultad de Ciencias Químicas, Unidad Saltillo, Universidad Autónoma de Coahuila, Boulevard Venustiano Carranza S/N Esquina Con Ing, José Cárdenas Valdés, República Oriente, Saltillo 25290, Mexico
| |
Collapse
|
30
|
Arnold N, Blaum C, Goßling A, Brunner FJ, Bay B, Zeller T, Ferrario MM, Brambilla P, Cesana G, Leoni V, Palmieri L, Donfrancesco C, Ojeda F, Linneberg A, Söderberg S, Iacoviello L, Gianfagna F, Costanzo S, Sans S, Veronesi G, Thorand B, Peters A, Tunstall-Pedoe H, Kee F, Salomaa V, Schnabel RB, Kuulasmaa K, Blankenberg S, Waldeyer C, Koenig W. Impact of Lipoprotein(a) Level on Low-Density Lipoprotein Cholesterol- or Apolipoprotein B-Related Risk of Coronary Heart Disease. J Am Coll Cardiol 2024; 84:165-177. [PMID: 38960510 DOI: 10.1016/j.jacc.2024.04.050] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Revised: 03/22/2024] [Accepted: 04/12/2024] [Indexed: 07/05/2024]
Abstract
BACKGROUND Conventional low-density lipoprotein cholesterol (LDL-C) quantification includes cholesterol attributable to lipoprotein(a) (Lp(a)-C) due to their overlapping densities. OBJECTIVES The purposes of this study were to compare the association between LDL-C and LDL-C corrected for Lp(a)-C (LDLLp(a)corr) with incident coronary heart disease (CHD) in the general population and to investigate whether concomitant Lp(a) values influence the association of LDL-C or apolipoprotein B (apoB) with coronary events. METHODS Among 68,748 CHD-free subjects at baseline LDLLp(a)corr was calculated as "LDL-C-Lp(a)-C," where Lp(a)-C was 30% or 17.3% of total Lp(a) mass. Fine and Gray competing risk-adjusted models were applied for the association between the outcome incident CHD and: 1) LDL-C and LDLLp(a)corr in the total sample; and 2) LDL-C and apoB after stratification by Lp(a) mass (≥/<90th percentile). RESULTS Similar risk estimates for incident CHD were found for LDL-C and LDL-CLp(a)corr30 or LDL-CLp(a)corr17.3 (subdistribution HR with 95% CI) were 2.73 (95% CI: 2.34-3.20) vs 2.51 (95% CI: 2.15-2.93) vs 2.64 (95% CI: 2.26-3.10), respectively (top vs bottom fifth; fully adjusted models). Categorization by Lp(a) mass resulted in higher subdistribution HRs for uncorrected LDL-C and incident CHD at Lp(a) ≥90th percentile (4.38 [95% CI: 2.08-9.22]) vs 2.60 [95% CI: 2.21-3.07]) at Lp(a) <90th percentile (top vs bottom fifth; Pinteraction0.39). In contrast, apoB risk estimates were lower in subjects with higher Lp(a) mass (2.43 [95% CI: 1.34-4.40]) than in Lp(a) <90th percentile (3.34 [95% CI: 2.78-4.01]) (Pinteraction0.49). CONCLUSIONS Correction of LDL-C for its Lp(a)-C content provided no meaningful information on CHD-risk estimation at the population level. Simple categorization of Lp(a) mass (≥/<90th percentile) influenced the association between LDL-C or apoB with future CHD mostly at higher Lp(a) levels.
Collapse
Affiliation(s)
- Natalie Arnold
- Department of Cardiology, University Heart & Vascular Center Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg, Germany; German Center for Cardiovascular Research (DZHK), partner site Hamburg/Kiel/Luebeck, Hamburg, Germany; Center for Population Health Innovation (POINT), University Heart and Vascular Center Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Christopher Blaum
- Department of Cardiology, University Heart & Vascular Center Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg, Germany; Center for Population Health Innovation (POINT), University Heart and Vascular Center Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Alina Goßling
- Department of Cardiology, University Heart & Vascular Center Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg, Germany; Center for Population Health Innovation (POINT), University Heart and Vascular Center Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Fabian J Brunner
- Department of Cardiology, University Heart & Vascular Center Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg, Germany; German Center for Cardiovascular Research (DZHK), partner site Hamburg/Kiel/Luebeck, Hamburg, Germany; Center for Population Health Innovation (POINT), University Heart and Vascular Center Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Benjamin Bay
- Department of Cardiology, University Heart & Vascular Center Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg, Germany; German Center for Cardiovascular Research (DZHK), partner site Hamburg/Kiel/Luebeck, Hamburg, Germany; Center for Population Health Innovation (POINT), University Heart and Vascular Center Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Tanja Zeller
- Department of Cardiology, University Heart & Vascular Center Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg, Germany; German Center for Cardiovascular Research (DZHK), partner site Hamburg/Kiel/Luebeck, Hamburg, Germany; Center for Population Health Innovation (POINT), University Heart and Vascular Center Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg, Germany; University Center of Cardiovascular Science at University Heart and Vascular Center Hamburg, Hamburg, Germany
| | - Marco M Ferrario
- Research Center in Epidemiology and Preventive Medicine - EPIMED, Department of Medicine and Surgery, University of Insubria, Varese, Italy
| | - Paolo Brambilla
- Department of Medicine and Surgery, University of Milano-Bicocca, Milan, Italy
| | - Giancarlo Cesana
- Department of Medicine and Surgery, University of Milano-Bicocca, Milan, Italy
| | - Valerio Leoni
- Laboratory of Clinical Pathology, Hospital Pio XI of Desio, ASST Brianza, School of Medicine and Surgery, University of Milano Bicocca, Milan, Italy
| | - Luigi Palmieri
- Department of Cardiovascular, Endocrine-Metabolic Diseases and Aging, Istituto Superiore di Sanità-ISS, Rome, Italy
| | - Chiara Donfrancesco
- Department of Cardiovascular, Endocrine-Metabolic Diseases and Aging, Istituto Superiore di Sanità-ISS, Rome, Italy
| | - Francisco Ojeda
- Department of Cardiology, University Heart & Vascular Center Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg, Germany; Center for Population Health Innovation (POINT), University Heart and Vascular Center Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Allan Linneberg
- Center for Clinical Research and Prevention, Bispebjerg and Frederiksberg Hospital, Copenhagen, Denmark; Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Stefan Söderberg
- Department of Public Health and Clinical Medicine, Umeå University, Umeå, Sweden
| | - Licia Iacoviello
- Department of Medicine and Surgery, LUM University, Casamassima, Italy; Department of Epidemiology and Prevention, IRCCS Neuromed, Pozzilli, Italy
| | - Francesco Gianfagna
- Research Center in Epidemiology and Preventive Medicine - EPIMED, Department of Medicine and Surgery, University of Insubria, Varese, Italy; Mediterranea Cardiocentro, Napoli, Italy
| | - Simona Costanzo
- Department of Epidemiology and Prevention, IRCCS Neuromed, Pozzilli, Italy
| | - Susana Sans
- Catalan Department of Health, Barcelona, Spain
| | - Giovanni Veronesi
- Research Center in Epidemiology and Preventive Medicine - EPIMED, Department of Medicine and Surgery, University of Insubria, Varese, Italy
| | - Barbara Thorand
- Institute of Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany; Institute for Medical Information Processing, Biometry, and Epidemiology-IBE, Ludwig-Maximilians University of Munich, Munich, Germany
| | - Annette Peters
- Institute of Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany; Institute for Medical Information Processing, Biometry, and Epidemiology-IBE, Ludwig-Maximilians University of Munich, Munich, Germany; German Center for Cardiovascular Disease Research (DZHK), partner site Munich Heart Alliance, Munich, Germany
| | - Hugh Tunstall-Pedoe
- Cardiovascular Epidemiology Unit, Institute of Cardiovascular Research, University of Dundee, Dundee, Scotland
| | - Frank Kee
- Centre for Public Health, Queens University of Belfast, Belfast, Northern Ireland, United Kingdom
| | - Veikko Salomaa
- Department of Public Health and Welfare, Finnish Institute for Health and Welfare (THL), Helsinki, Finland
| | - Renate B Schnabel
- Department of Cardiology, University Heart & Vascular Center Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg, Germany; German Center for Cardiovascular Research (DZHK), partner site Hamburg/Kiel/Luebeck, Hamburg, Germany; Center for Population Health Innovation (POINT), University Heart and Vascular Center Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Kari Kuulasmaa
- Department of Public Health and Welfare, Finnish Institute for Health and Welfare (THL), Helsinki, Finland
| | - Stefan Blankenberg
- Department of Cardiology, University Heart & Vascular Center Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg, Germany; German Center for Cardiovascular Research (DZHK), partner site Hamburg/Kiel/Luebeck, Hamburg, Germany; Center for Population Health Innovation (POINT), University Heart and Vascular Center Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Christoph Waldeyer
- Department of Cardiology, University Heart & Vascular Center Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg, Germany; German Center for Cardiovascular Research (DZHK), partner site Hamburg/Kiel/Luebeck, Hamburg, Germany; Center for Population Health Innovation (POINT), University Heart and Vascular Center Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Wolfgang Koenig
- German Heart Center, Munich, Technical University of Munich, Munich, Germany; Institute of Epidemiology and Medical Biometry, University of Ulm, Ulm, Germany; German Center for Cardiovascular Disease Research (DZHK), partner site Munich Heart Alliance, Munich, Germany.
| |
Collapse
|
31
|
Brandt EJ, Kirch M, Patel N, Chennareddy C, Murthy VL, Goonewardena SN. Impact of Social Determinants of Health and Lifestyle on Association Between Lipoprotein(a) and Cardiovascular Events. JACC. ADVANCES 2024; 3:101016. [PMID: 39129977 PMCID: PMC11313040 DOI: 10.1016/j.jacadv.2024.101016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Revised: 04/11/2024] [Accepted: 04/18/2024] [Indexed: 08/13/2024]
Abstract
Background In European cohorts, healthier lifestyle either attenuated or associated with lower cardiovascular risk despite elevated lipoprotein(a) [Lp(a)]. Objectives The purpose of this study was to test if social determinants of health (SDOH) and Life's Simple 7 (LS7) scores impact the association of Lp(a) with cardiovascular events in U.S. cohorts. Methods We performed a sequential multivariable Cox proportional hazard analysis using the ARIC (Atherosclerosis Risk In Communities) and MESA (Multi-Ethnic Study of Atherosclerosis) cohorts. We first adjusted for age, gender, non-high-density lipoprotein-cholesterol, race, and ethnicity, then sequentially added SDOH and LS7 scores. The primary outcomes were time until first myocardial infarction (MI) or stroke. Results ARIC (n = 15,072; median Lp(a) = 17.3 mg/dL) had 16.2 years and MESA (n = 6,822; median Lp(a) = 18.3 mg/dL) had 12.3 years of average follow-up. In age, gender, race, and ethnicity, and non-high-density lipoprotein-cholesterol adjusted analyses, Lp(a) was associated with MI in ARIC (HR: 1.10, P < 0.001) and MESA (HR: 1.11, P = 0.001), and stroke in ARIC (HR: 1.07, P < 0.001) but not MESA (HR: 0.97, P = 0.53). In models with SDOH and LS7, associations of Lp(a) remained similar with MI (ARIC, HR: 1.08, P < 0.001; MESA, HR: 1.10, P = 0.001) and stroke (ARIC, HR: 1.06, P = 0.002; MESA, HR: 0.96, P = 0.37). Each additional SDOH correlated positively with MI (ARIC, HR: 1.04, P = 0.01; MESA, HR: 1.08, P = 0.003) and stroke in ARIC (HR: 1.08, P = 0.00) but not MESA (HR: 1.03, P = 0.41). Each additional LS7 point correlated negatively with MI (ARIC, HR: 0.88, P < 0.001; MESA, HR: 0.85, P < 0.001) and stroke (ARIC, HR: 0.91, P < 0.001; MESA, HR: 0.86, P < 0.001). Conclusions SDOH and lifestyle factors associated with risk for MI and stroke but did not largely impact the association between Lp(a) and cardiovascular events.
Collapse
Affiliation(s)
- Eric J. Brandt
- Institute for Healthcare Policy and Innovation, University of Michigan, Ann Arbor, Michigan, USA
- Division of Cardiovascular Medicine, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Matthias Kirch
- Institute for Healthcare Policy and Innovation, University of Michigan, Ann Arbor, Michigan, USA
| | - Nimai Patel
- Division of Cardiovascular Medicine, Department of Internal Medicine, The University of Texas Health Science Center at Houston, Houston, Texas, USA
| | | | - Venkatesh L. Murthy
- Division of Cardiovascular Medicine, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
- Divisions of Nuclear Medicine and Cardiothoracic Imaging, Department of Radiology, University of Michigan, Ann Arbor, Michigan, USA
| | - Sascha N. Goonewardena
- Division of Cardiovascular Medicine, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| |
Collapse
|
32
|
Tsimikas S. Lipoprotein(a) in the Year 2024: A Look Back and a Look Ahead. Arterioscler Thromb Vasc Biol 2024; 44:1485-1490. [PMID: 38924439 PMCID: PMC11210685 DOI: 10.1161/atvbaha.124.319483] [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] [Indexed: 06/28/2024]
Abstract
In fitting with the American Heart Association’s 100th anniversary of its founding and Arteriosclerosis, Thrombosis and Vascular Biology organizing a Centennial Collection to celebrate this event, lipoprotein(a) [Lp(a)] celebrates its 61st birthday in November 2024. There has been substantial progress in understanding the biology and pathophysiology of Lp(a) in the last 6 decades, including its discovery as a unique β-lipoprotein containing the pathognomonic apolipoprotein(a) moiety covalently bound to apolipoprotein B-100, its independent monogenetic association with cardiovascular disease and calcific aortic valve disease, its increased content of pro-atherogenic and pro-inflammatory of oxidized phospholipids relative to other lipoproteins and the development of RNA therapeutics to lower plasma Lp(a) levels. The validation or refutation of the “Lp(a) hypothesis”, namely that lowering plasma Lp(a) will lead to clinical benefit, is ongoing in 3 clinical outcomes trials. This essay reviews the discovery of Lp(a), summarizes the seminal pathophysiological findings since its discovery, discusses ongoing clinical trials with novel drugs and approaches, and provides a look ahead to unanswered questions.
Collapse
|
33
|
Bellomo TR, Liu Y, Gilliland TC, Miksenas H, Haidermota S, Wong M, Hu X, Cristino JR, Browne A, Plutzky J, Tsimikas S, Januzzi JL, Natarajan P. Associations between lipoprotein(a), oxidized phospholipids, and extracoronary vascular disease. J Lipid Res 2024; 65:100585. [PMID: 38942114 PMCID: PMC11298641 DOI: 10.1016/j.jlr.2024.100585] [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: 03/04/2024] [Revised: 06/20/2024] [Accepted: 06/21/2024] [Indexed: 06/30/2024] Open
Abstract
The roles of lipoprotein(a) [Lp(a)] and related oxidized phospholipids (OxPLs) in the development and progression of coronary disease is known, but their influence on extracoronary vascular disease is not well-established. We sought to evaluate associations between Lp(a), OxPL apolipoprotein B (OxPL-apoB), and apolipoprotein(a) (OxPL-apo(a)) with angiographic extracoronary vascular disease and incident major adverse limb events (MALEs). Four hundred forty-six participants who underwent coronary and/or peripheral angiography were followed up for a median of 3.7 years. Lp(a) and OxPLs were measured before angiography. Elevated Lp(a) was defined as ≥150 nmol/L. Elevated OxPL-apoB and OxPL-apo(a) were defined as greater than or equal to the 75th percentile (OxPL-apoB ≥8.2 nmol/L and OxPL-apo(a) ≥35.8 nmol/L, respectively). Elevated Lp(a) had a stronger association with the presence of extracoronary vascular disease compared to OxPLs and was minimally improved with the addition of OxPLs in multivariable models. Compared to participants with normal Lp(a) and OxPL concentrations, participants with elevated Lp(a) levels were twice as likely to experience a MALE (odds ratio: 2.14, 95% confidence interval: 1.03, 4.44), and the strength of the association as well as the C statistic of 0.82 was largely unchanged with the addition of OxPL-apoB and OxPL-apo(a). Elevated Lp(a) and OxPLs are risk factors for progression and complications of extracoronary vascular disease. However, the addition of OxPLs to Lp(a) does not provide additional information about risk of extracoronary vascular disease. Therefore, Lp(a) alone captures the risk profile of Lp(a), OxPL-apoB, and OxPL-apo(a) in the development and progression of atherosclerotic plaque in peripheral arteries.
Collapse
Affiliation(s)
- Tiffany R Bellomo
- Division of Vascular and Endovascular Surgery, Massachusetts General Hospital, Boston, MA, USA
| | - Yuxi Liu
- Division of Cardiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Thomas C Gilliland
- Division of Cardiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA; Program in Medical and Population Genetics and the Cardiovascular Disease Initiative, Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Hannah Miksenas
- Division of Cardiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Sara Haidermota
- Division of Cardiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Megan Wong
- Division of Cardiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Xingdi Hu
- Novartis Pharmaceuticals Corporation, Novartis, East Hanover, NJ, USA
| | | | - Auris Browne
- Novartis Pharmaceuticals Corporation, Novartis, East Hanover, NJ, USA
| | - Jorge Plutzky
- Division of Cardiology, Brigham and Women's Hospital, Boston, Harvard Medical School, Boston, MA, USA
| | - Sotirios Tsimikas
- Sulpizio Cardiovascular Center, University of California San Diego, La Jolla, CA, USA
| | - James L Januzzi
- Division of Cardiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA; Cardiology Division, Baim Institute for Clinical Research, Boston, MA, USA
| | - Pradeep Natarajan
- Division of Cardiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA; Program in Medical and Population Genetics and the Cardiovascular Disease Initiative, Broad Institute of Harvard and MIT, Cambridge, MA, USA.
| |
Collapse
|
34
|
Januzzi JL, van Kimmenade RRJ, Liu Y, Hu X, Browne A, Plutzky J, Tsimikas S, Blankstein R, Natarajan P. Lipoprotein(a), Oxidized Phospholipids, and Progression to Symptomatic Heart Failure: The CASABLANCA Study. J Am Heart Assoc 2024; 13:e034774. [PMID: 38860394 PMCID: PMC11255745 DOI: 10.1161/jaha.124.034774] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Accepted: 05/09/2024] [Indexed: 06/12/2024]
Abstract
BACKGROUND Higher lipoprotein(a) and oxidized phospholipid concentrations are associated with increased risk for coronary artery disease and valvular heart disease. The role of lipoprotein(a) or oxidized phospholipid as a risk factor for incident heart failure (HF) or its complications remains uncertain. METHODS AND RESULTS A total of 1251 individuals referred for coronary angiography in the Catheter Sampled Blood Archive in Cardiovascular Diseases (CASABLANCA) study were stratified on the basis of universal definition of HF stage; those in stage A/B (N=714) were followed up for an average 3.7 years for incident stage C/D HF or the composite of HF/cardiovascular death. During follow-up, 105 (14.7%) study participants in stage A/B progressed to symptomatic HF and 57 (8.0%) had cardiovascular death. In models adjusted for multiple HF risk factors, including severe coronary artery disease and aortic stenosis, individuals with lipoprotein(a) ≥150 nmol/L were at higher risk for progression to symptomatic HF (hazard ratio [HR], 1.90 [95% CI, 1.15-3.13]; P=0.01) or the composite of HF/cardiovascular death (HR, 1.71 [95% CI, 1.10-2.67]; P=0.02). These results remained significant after further adjustment of the model to include prior myocardial infarction (HF: HR, 1.89, P=0.01; HF/cardiovascular death: HR, 1.68, P=0.02). Elevated oxidized phospholipid concentrations were similarly associated with risk, particularly when added to higher lipoprotein(a). In Kaplan-Meier analyses, individuals with stage A/B HF and elevated lipoprotein(a) had shorter time to progression to stage C/D HF or HF/cardiovascular death (both log-rank P<0.001). CONCLUSIONS Among individuals with stage A or B HF, higher lipoprotein(a) and oxidized phospholipid concentrations are independent risk factors for progression to symptomatic HF or cardiovascular death. REGISTRATION URL: https://wwwclinicaltrials.gov; Unique identifier: NCT00842868.
Collapse
Affiliation(s)
- James L. Januzzi
- Division of CardiologyMassachusetts General Hospital, Harvard Medical SchoolBostonMA
- Baim Institute for Clinical ResearchBostonMA
| | | | - Yuxi Liu
- Division of CardiologyMassachusetts General Hospital, Harvard Medical SchoolBostonMA
| | - Xingdi Hu
- Novartis Pharmaceuticals CorporationEast HanoverNJ
| | - Auris Browne
- Novartis Pharmaceuticals CorporationEast HanoverNJ
| | - Jorge Plutzky
- Cardiovascular DivisionBrigham and Women’s Hospital, Harvard Medical SchoolBostonMA
| | - Sotirios Tsimikas
- Sulpizio Cardiovascular CenterUniversity of California San DiegoLa JollaCA
| | - Ron Blankstein
- Cardiovascular DivisionBrigham and Women’s Hospital, Harvard Medical SchoolBostonMA
| | - Pradeep Natarajan
- Division of CardiologyMassachusetts General Hospital, Harvard Medical SchoolBostonMA
- Program in Medical and Population Genetics and the Cardiovascular Disease InitiativeBroad Institute of Harvard and MITCambridgeMA
| |
Collapse
|
35
|
Wang Y, Li X, Huang R, Chen XW, Wang X. Apolipoprotein B Secretion Assay from Primary Hepatocytes. Bio Protoc 2024; 14:e4982. [PMID: 38737509 PMCID: PMC11082783 DOI: 10.21769/bioprotoc.4982] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Revised: 03/27/2024] [Accepted: 04/01/2024] [Indexed: 05/14/2024] Open
Abstract
Apolipoprotein B (APOB) is the primary structural protein of atherogenic lipoproteins, which drive atherogenesis and thereby lead to deadly cardiovascular diseases (CVDs). Plasma levels of APOB-containing lipoproteins are tightly modulated by LDL receptor-mediated endocytic trafficking and cargo receptor-initiated exocytic route; the latter is much less well understood. This protocol aims to present an uncomplicated yet effective method for detecting APOB/lipoprotein secretion. We perform primary mouse hepatocyte isolation and culture coupled with well-established techniques such as immunoblotting for highly sensitive, specific, and semi-quantitative analysis of the lipoprotein secretion process. Its inherent simplicity facilitates ease of operation, rendering it a valuable tool widely utilized to explore the intricate landscape of cellular lipid metabolism and unravel the mechanistic complexities underlying lipoprotein-related diseases. Key features • A pipeline for the isolation and subsequent culture of mouse primary hepatocytes. • A procedure for tracking the secretion of APOB-containing lipoproteins. • A rapid and sensitive assay for detecting the APOB level based on immunoblotting.
Collapse
Affiliation(s)
- Yawei Wang
- State Key Laboratory of Membrane Biology, Peking University, Beijing, China
- PKU-THU Joint Center for Life Sciences, Peking University, Beijing, China
| | - Xin Li
- State Key Laboratory of Membrane Biology, Peking University, Beijing, China
- College of Future Technology, Institute of Molecular Medicine, Peking University, Beijing, China
| | - Runze Huang
- State Key Laboratory of Membrane Biology, Peking University, Beijing, China
- College of Future Technology, Institute of Molecular Medicine, Peking University, Beijing, China
| | - Xiao-Wei Chen
- State Key Laboratory of Membrane Biology, Peking University, Beijing, China
- PKU-THU Joint Center for Life Sciences, Peking University, Beijing, China
- College of Future Technology, Institute of Molecular Medicine, Peking University, Beijing, China
| | - Xiao Wang
- State Key Laboratory of Membrane Biology, Peking University, Beijing, China
- PKU-THU Joint Center for Life Sciences, Peking University, Beijing, China
- College of Future Technology, Institute of Molecular Medicine, Peking University, Beijing, China
| |
Collapse
|
36
|
Bellinge JW, Chan DC, Pang J, Francis RJ, Page MM, Watts GF, Schultz CJ. Plasma lipoprotein(a) is associated with calcification activity of the thoracic aorta and aortic valve in statin naïve individuals with diabetes mellitus. Eur J Clin Invest 2024; 54:e14167. [PMID: 38265272 DOI: 10.1111/eci.14167] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Revised: 01/11/2024] [Accepted: 01/12/2024] [Indexed: 01/25/2024]
Affiliation(s)
- Jamie W Bellinge
- Medical School, University of Western Australia, Perth, Western Australia, Australia
- Department of Nuclear Medicine, Sir Charles Gairdner Hospital, Nedlands, Western Australia, Australia
| | - Dick C Chan
- Medical School, University of Western Australia, Perth, Western Australia, Australia
| | - Jing Pang
- Medical School, University of Western Australia, Perth, Western Australia, Australia
| | - Roslyn J Francis
- Medical School, University of Western Australia, Perth, Western Australia, Australia
- Department of Nuclear Medicine, Sir Charles Gairdner Hospital, Nedlands, Western Australia, Australia
| | - Michael M Page
- Medical School, University of Western Australia, Perth, Western Australia, Australia
- Department of Clinical Biochemistry, Western Diagnostic Pathology, Perth, Australia
| | - Gerald F Watts
- Medical School, University of Western Australia, Perth, Western Australia, Australia
- Cardiometabolic Service, Department of Cardiology, Royal Perth Hospital, Perth, Western Australia, Australia
| | - Carl J Schultz
- Medical School, University of Western Australia, Perth, Western Australia, Australia
- Department of Cardiology, Royal Perth Hospital, Perth, Western Australia, Australia
| |
Collapse
|
37
|
Tsimikas S, Bittner V. Particle Number and Characteristics of Lipoprotein(a), LDL, and apoB: Perspectives on Contributions to ASCVD. J Am Coll Cardiol 2024; 83:396-400. [PMID: 38233013 DOI: 10.1016/j.jacc.2023.11.008] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Accepted: 11/08/2023] [Indexed: 01/19/2024]
Affiliation(s)
| | - Vera Bittner
- Division of Cardiovascular Disease, University of Alabama at Birmingham, Birmingham, Alabama, USA
| |
Collapse
|
38
|
Hooper AJ, Fernando PMS, Burnett JR. Potential of muvalaplin as a lipoprotein(a) inhibitor. Expert Opin Investig Drugs 2024; 33:5-7. [PMID: 38186354 DOI: 10.1080/13543784.2024.2302592] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Accepted: 01/03/2024] [Indexed: 01/09/2024]
Affiliation(s)
- Amanda J Hooper
- Department of Clinical Biochemistry, PathWest Laboratory Medicine WA, Royal Perth Hospital & Fiona Stanley Hospital Network, Perth, Western Australia, Australia
- School of Medicine, University of Western Australia, Perth, Western Australia, Australia
| | - P Mihika S Fernando
- Department of Clinical Biochemistry, PathWest Laboratory Medicine WA, Royal Perth Hospital & Fiona Stanley Hospital Network, Perth, Western Australia, Australia
| | - John R Burnett
- Department of Clinical Biochemistry, PathWest Laboratory Medicine WA, Royal Perth Hospital & Fiona Stanley Hospital Network, Perth, Western Australia, Australia
- School of Medicine, University of Western Australia, Perth, Western Australia, Australia
| |
Collapse
|
39
|
Hajeyah AA, Protty MB, Paul D, Costa D, Omidvar N, Morgan B, Iwasaki Y, McGill B, Jenkins PV, Yousef Z, Allen-Redpath K, Soyama S, Choudhury A, Mitra R, Yaqoob P, Morrissey JH, Collins PW, O'Donnell VB. Phosphatidylthreonine is a procoagulant lipid detected in human blood and elevated in coronary artery disease. J Lipid Res 2024; 65:100484. [PMID: 38103786 PMCID: PMC10809103 DOI: 10.1016/j.jlr.2023.100484] [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: 06/19/2023] [Revised: 12/11/2023] [Accepted: 12/11/2023] [Indexed: 12/19/2023] Open
Abstract
Aminophospholipids (aPL) such as phosphatidylserine are essential for supporting the activity of coagulation factors, circulating platelets, and blood cells. Phosphatidylthreonine (PT) is an aminophospholipid previously reported in eukaryotic parasites and animal cell cultures, but not yet in human tissues. Here, we evaluated whether PT is present in blood cells and characterized its ability to support coagulation. Several PT molecular species were detected in human blood, washed platelets, extracellular vesicles, and isolated leukocytes from healthy volunteers using liquid chromatography-tandem mass spectrometry. The ability of PT to support coagulation was demonstrated in vitro using biochemical and biophysical assays. In liposomes, PT supported prothrombinase activity in the presence and absence of phosphatidylserine. PT nanodiscs strongly bound FVa and lactadherin (nM affinity) but poorly bound prothrombin and FX, suggesting that PT supports prothrombinase through recruitment of FVa. PT liposomes bearing tissue factor poorly generated thrombin in platelet poor plasma, indicating that PT poorly supports extrinsic tenase activity. On platelet activation, PT is externalized and partially metabolized. Last, PT was significantly higher in platelets and extracellular vesicle from patients with coronary artery disease than in healthy controls. In summary, PT is present in human blood, binds FVa and lactadherin, supports coagulation in vitro through FVa binding, and is elevated in atherosclerotic vascular disease. Our studies reveal a new phospholipid subclass, that contributes to the procoagulant membrane, and may support thrombosis in patients at elevated risk.
Collapse
Affiliation(s)
- Ali A Hajeyah
- Systems Immunity Research Institute and Division of Infection and Immunity, Cardiff University, Cardiff, United Kingdom; Department of Biological Sciences, Kuwait University, Safat, Kuwait.
| | - Majd B Protty
- Systems Immunity Research Institute and Division of Infection and Immunity, Cardiff University, Cardiff, United Kingdom
| | - Divyani Paul
- Department of Biological Chemistry, University of Michigan, Ann Arbor, MI, USA
| | - Daniela Costa
- Systems Immunity Research Institute and Division of Infection and Immunity, Cardiff University, Cardiff, United Kingdom
| | - Nader Omidvar
- Systems Immunity Research Institute and Division of Infection and Immunity, Cardiff University, Cardiff, United Kingdom
| | - Bethan Morgan
- Systems Immunity Research Institute and Division of Infection and Immunity, Cardiff University, Cardiff, United Kingdom
| | - Yugo Iwasaki
- College of Bioscience and Biotechnology, Chubu University, Kasugai, Japan
| | - Beth McGill
- Systems Immunity Research Institute and Division of Infection and Immunity, Cardiff University, Cardiff, United Kingdom
| | | | - Zaheer Yousef
- University Hospital of Wales, Cardiff, United Kingdom
| | - Keith Allen-Redpath
- Department of Food and Nutritional Sciences, University of Reading, Reading, United Kingdom
| | - Shin Soyama
- Department of Food and Nutritional Sciences, University of Reading, Reading, United Kingdom
| | | | - Rito Mitra
- University Hospital of Wales, Cardiff, United Kingdom
| | - Parveen Yaqoob
- Department of Food and Nutritional Sciences, University of Reading, Reading, United Kingdom
| | - James H Morrissey
- Department of Biological Chemistry, University of Michigan, Ann Arbor, MI, USA
| | - Peter W Collins
- Systems Immunity Research Institute and Division of Infection and Immunity, Cardiff University, Cardiff, United Kingdom; University Hospital of Wales, Cardiff, United Kingdom
| | - Valerie B O'Donnell
- Systems Immunity Research Institute and Division of Infection and Immunity, Cardiff University, Cardiff, United Kingdom.
| |
Collapse
|