1
|
Athauda-Arachchi PM, Kaththiriarachchi L, Salgado W, De Silva S, Salgado T, Farooq M, Jibran MJ, Godakanda Arachchi Y, Premanath S, Senaratne SL, Samarakoon SP, Rathnayake H, Shajahan M. Cross-sectional study of lipoprotein(a) and the severity of coronary artery disease, cerebrovascular disease, and peripheral vascular disease in a group of South Asian patients. Cardiovasc Endocrinol Metab 2025; 14:e00327. [PMID: 40160971 PMCID: PMC11952828 DOI: 10.1097/xce.0000000000000327] [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: 01/09/2025] [Accepted: 02/27/2025] [Indexed: 04/02/2025]
Abstract
Background Atherosclerotic cardiovascular diseases (ASCVD), including coronary artery disease (CAD), cerebrovascular disease (CVD), and peripheral vascular disease (PVD), remain the leading cause of death globally. South Asians exhibit a higher incidence of cardiovascular diseases than other ethnicities, attributed to a range of genetic, environmental, and lifestyle factors. Lipoprotein(a) [Lp(a)] with a unique apolipoprotein(a) component, has emerged as a marker of atherosclerosis and ASCVD risk, with evidence to promote arterial plaque formation and thrombogenesis. Objective The aim of this study was to explore the associations between Lp(a) levels and the severity of CAD, CVD, and PVD in a group of South Asian patients. Methods Following ethical approval, 60 consecutive patients who underwent coronary angiography for any indication were reviewed. There were 51 eligible participants who were evaluated for Lp(a) level, Synergy Between Percutaneous Coronary Intervention with Taxus and Cardiac Surgery (SYNTAX) score, and severity of CVD and PVD. The SYNTAX-I score was calculated using two-observer consensus on coronary angiograms. Assessment of CVD was by ultrasound/Doppler, and PVD by estimating ankle-brachial index using Doppler. The multisite arterial disease score 2 (MADS2) and SYNTAX score tertiles were used to group the patients. Statistical analysis was performed using the SPSS software. Results and discussion In this group, we identified a statistically significant difference with higher Lp(a) levels being associated with more severe coronary disease (SYNTAX tertile 2,3). Despite a numerical trend, statistical significance was not confirmed for Lp(a) levels in relation to MADS2-CVD or MADS2-PVD scores. A larger study may be required to assess these aspects.
Collapse
|
2
|
Liu S, Wang X, Hu J, Zhao C, Qin X. Comprehensive evaluation of siRNA therapeutics on Lp(a): A network meta-analysis. Diabetes Obes Metab 2025; 27:3367-3378. [PMID: 40116213 DOI: 10.1111/dom.16355] [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: 01/18/2025] [Revised: 03/02/2025] [Accepted: 03/09/2025] [Indexed: 03/23/2025]
Abstract
AIMS To evaluate the efficacy and safety of siRNA drugs that lower Lp(a) in patients with dyslipidaemia. MATERIALS AND METHODS A network meta-analysis and systematic review were conducted to compare siRNA drugs targeting Lp(a), based on relevant randomized controlled trials (RCTs). A comprehensive search was performed in PubMed, Embase, Web of Science and the Cochrane Library (up to October 24, 2024). RCTs with an intervention duration of at least 12 weeks were included. Eligible studies compared siRNA drugs that reduce Lp(a), including both Lp(a)-targeted and non-targeted agents, with placebo or other siRNA drugs that reduce Lp(a). The primary outcomes were the percentage reduction and absolute reduction in Lp(a), percentage reduction in low-density lipoprotein cholesterol (LDL-C), percentage reduction in apolipoprotein B (apo(B)), adverse events and serious adverse events, including injection-site reactions. The risk of bias was assessed using the Cochrane Risk of Bias Tool (ROB2), and a random-effects network meta-analysis was performed using the frequentist approach. Confidence in effect estimates was evaluated using the Confidence In Network Meta-Analysis (CINeMA) framework. RESULTS A total of 14 trials involving 5646 participants were included. Lp(a)-targeted siRNA agents, particularly Olpasiran, demonstrated strong efficacy in significantly reducing Lp(a) levels, with the greatest percentage reduction in Lp(a) (mean difference [MD]: -92.06%; 95% CI: -102.43% to -81.69%; P-score: 0.98). Olpasiran also showed the greatest absolute reduction in Lp(a) (MD: -250.70 nmol/L; 95% confidence interval [CI]: -279.89 to -221.50; P-score: 0.99). Certain non-Lp(a)-targeted siRNA agents, such as inclisiran and zodasiran, also showed modest reductions in Lp(a) levels, reducing Lp(a) by approximately 15%. Lp(a)-targeted siRNA agents reduced LDL-C by more than 20% and decreased apo(B) by approximately 15%. In terms of safety, most drugs exhibited favourable safety profiles with no significant differences compared to placebo. However, zerlasiran raised concerns regarding injection-site reactions and other adverse events when compared to placebo. CONCLUSIONS Lp(a)-targeted siRNA agents have shown robust effectiveness in substantially reducing Lp(a) levels, including both percentage and absolute reductions, with moderate improvements in LDL-C and apo(B) concentrations. Non-Lp(a)-targeted siRNA agents also demonstrate modest reductions in Lp(a) levels. The safety profile is generally favourable, but zerlasiran and inclisiran may increase the incidence of injection-site reactions.
Collapse
Affiliation(s)
- Song Liu
- Department of Pharmacy, The Third People's Hospital of Chengdu, The Affiliated Hospital of Southwest Jiaotong University, Chengdu, Sichuan, China
| | - Xingjin Wang
- Medical Faculty, Heidelberg University, Heidelberg, Germany
| | - Jiaqiang Hu
- Department of Pharmacy, Guang'an People's Hospital, Guang'an, Sichuan, China
| | - Chen Zhao
- Department of Pharmacy, Beijing Anzhen Nanchong Hospital, Capital Medical University & Nanchong Central Hospital, Nanchong, Sichuan, China
- Nanchong Key Laboratory of Individualized Drug Therapy, Nanchong, China
| | - Xiaoli Qin
- Department of Pharmacy, The Third People's Hospital of Chengdu, The Affiliated Hospital of Southwest Jiaotong University, Chengdu, Sichuan, China
| |
Collapse
|
3
|
Xu R, Wang Z, Dong J, Yu M, Zhou Y. Lipoprotein(a) and panvascular disease. Lipids Health Dis 2025; 24:186. [PMID: 40413492 DOI: 10.1186/s12944-025-02600-y] [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: 02/17/2025] [Accepted: 05/08/2025] [Indexed: 05/27/2025] Open
Abstract
Panvascular disease (PVD) is an emerging clinical concept that encompasses a spectrum of atherosclerotic conditions involving multiple major vascular beds, including the coronary, cerebral, peripheral, and valvular arteries. Although not formally recognized as a nosological entity, in this review, PVD is adopted as a conceptual framework to reflect the systemic nature of atherosclerosis affecting vascular territories supplying the heart, brain, and peripheral circulation. This perspective enables a more integrated understanding of disease processes across organ systems that are often studied in isolation. Lipoprotein(a) [Lp(a)] is a genetically regulated, low-density lipoprotein (LDL)-like particle that has garnered increasing attention as an independent pathogenic risk factor for PVD. Accumulating evidence from epidemiological, genetic, and mechanistic studies has confirmed the multifaceted role of Lp(a) in promoting atherogenesis, vascular calcification, inflammation, and thrombogenesis across multiple vascular beds. Elevated Lp(a) levels are associated with increased cardiovascular and cerebrovascular event risk, even after controlling for traditional risk factors. This review systematically outlines the structure, genetic determinants, and pathogenic mechanisms of Lp(a), and synthesizes current clinical evidence regarding its role in various PVD subtypes. The interactions between Lp(a) and traditional cardiovascular risk factors such as hypercholesterolemia, diabetes, and hypertension are explored in depth, highlighting their synergistic contributions to vascular injury and disease progression. Furthermore, sex-based differences in Lp(a)-associated risk, response to therapy, and biological behavior are discussed, providing insights into personalized cardiovascular risk stratification. In addition, the review summarizes current and emerging therapeutic strategies targeting Lp(a), including niacin, antisense oligonucleotides (ASOs), small interfering RNAs (siRNAs), and gene-editing technologies. These advances offer promising new avenues for reducing residual cardiovascular risk attributable to elevated Lp(a). In conclusion, viewing Lp(a)-associated pathology through the lens of PVD provides a comprehensive and unifying approach to understanding its systemic impact. This framework supports the development of integrated risk assessment tools and multi-targeted interventions, ultimately aiming to improve outcomes for patients with complex, multisite vascular involvement.
Collapse
Affiliation(s)
- Ruiyan Xu
- Department of Clinical Medicine, Queen Mary School of Nanchang University, Nanchang, 330031, China
| | - Zhenwei Wang
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450000, China
| | - Jiayu Dong
- Department of Clinical Medicine, Queen Mary School of Nanchang University, Nanchang, 330031, China
| | - Miao Yu
- Department of Cardiovascular Medicine, The Second Affiliated Hospital of Nanchang University, Nanchang, 330008, China.
| | - Yue Zhou
- Department of Cardiovascular Medicine, The Second Affiliated Hospital of Nanchang University, Nanchang, 330008, China.
| |
Collapse
|
4
|
Liu Y, Li M, Zhang H, Gao L, Liu J, Hou Y, Xu J. Development of a fully automated latex-enhanced immunoturbidimetric method for quantitative serum Lp(a) measurement. Biotechnol Lett 2025; 47:31. [PMID: 40045089 DOI: 10.1007/s10529-025-03564-w] [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: 06/08/2024] [Revised: 01/13/2025] [Accepted: 01/20/2025] [Indexed: 05/13/2025]
Abstract
BACKGROUND Lipoprotein (a) [Lp(a)] is a critical factor in cardiovascular health, composed of low-density lipoprotein-like particles bound to apolipoprotein (a). Elevated Lp(a) levels are associated with an increased risk of cardiovascular diseases (CVD), accelerating disease progression and raising CVD-related mortality. However, the lack of standardized measurement methods for Lp(a) contributes to diagnostic uncertainties in this area. METHOD A quantitative measurement method for serum Lp(a) was developed using fully automated latex-enhanced particle immunoturbidimetry, marking a significant advancement in diagnostic capabilities. Key parameters, including repeatability, stability, linearity, detection limit, interference, and method comparison, were evaluated to ensure the assay's reliability and accuracy. RESULT Lp(a) in samples was detected by carboxylated latex particles (95 nm in diameter) covalently coated with anti-Lp(a) antibodies. Lp(a) concentration was quantified by measuring the turbidity changes caused by agglutination at 600 nm. This method provides rapid, accurate, and fully automated measurements on the Hitachi 7100 automatic biochemical analyzer. With intra-batch precision CV% of 1.10% and inter-batch precision CV% of 1.79%, the method demonstrates reliable performance with Randox biochemical quality control samples. It has a detection limit of 7 mg/L and a high correlation coefficient (R2 = 0.9946) within the 0-1500 mg/L range. Minimal interference from bilirubin, fat emulsion, hemoglobin, and ascorbic acid was observed. Additionally, it shows strong correlation (R2 = 0.9972) with a commercially available latex-enhanced immunoturbidimetric Lp(a) assay reagent, confirming its comparability and clinical suitability. CONCLUSION The quantitative serum Lp(a) determination method based on latex-enhanced immunoturbidimetry offers numerous advantages. It provides rapid, accurate, and automated results, making it ideal for routine clinical testing. The method effectively measures Lp(a) in serum samples by leveraging the interaction between Lp(a) and latex particles.
Collapse
Affiliation(s)
- Yanyan Liu
- School of Life Science and Technology, Changchun University of Science and Technology, Changchun, 130013, Jilin, P.R. China
| | - Meijiao Li
- School of Life Science and Technology, Changchun University of Science and Technology, Changchun, 130013, Jilin, P.R. China
| | - Hao Zhang
- School of Life Science and Technology, Changchun University of Science and Technology, Changchun, 130013, Jilin, P.R. China
| | - Le Gao
- School of Life Science and Technology, Changchun University of Science and Technology, Changchun, 130013, Jilin, P.R. China
| | - Jitao Liu
- Jilin Getein Biotechnology Co., Ltd., Changchun, 130103, Jilin, P.R. China
| | - Yue Hou
- School of Life Science and Technology, Changchun University of Science and Technology, Changchun, 130013, Jilin, P.R. China.
| | - Jiancheng Xu
- Department of Laboratory Medicine, First Hospital of Jilin University, No. 1 Xinmin Street, Changchun, 130021, P.R. China.
| |
Collapse
|
5
|
Velilla TA, Prendes CF, Grau NA, Calmarza P, Anguila SC, Estébanez BC, Castro MJC, Ceacero D, Martínez IG, Palencia MM, Foncillas JP, Román CR, on behalf of the SEQC ML Working Group on Lipoproteins and Cardiovascular Diseases. Fundamentals of lipoprotein(a) request and quantification in the clinical laboratory. ADVANCES IN LABORATORY MEDICINE 2025; 6:7-16. [PMID: 40160391 PMCID: PMC11949557 DOI: 10.1515/almed-2025-0034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Accepted: 01/08/2025] [Indexed: 04/02/2025]
Abstract
Cardiovascular diseases keep being the leading cause of mortality in Spain. Efforts should be intensified to identify new risk factors that may contribute to increasing cardiovascular risk. Lipoprotein(a) (Lp(a)) has been associated with a higher risk for developing aortic valve stenosis, heart failure, ischemic stroke, ischemic heart disease and peripheral arterial disease. Hyperlipoproteinemia(a) is a common health problem. Between 10 and 30 % of the world population have Lp(a) values exceeding 50 mg/dL. The scientific evidence provided in the recent years confirms an independent association between Lp(a) and the risk for having an arteriosclerotic cardiovascular event. This finding, added to the emergence of new specific therapies for reducing Lp(a) has raised interest in the quantification of this lipoprotein. The objective of this paper was to perform a review of the evidence available to identify the patients who will benefit from undergoing Lp(a) testing and determine the recommended quantification methods, the desirable concentrations, and the role of Lp(a) determination in reclassifying the cardiovascular risk of patients.
Collapse
Affiliation(s)
| | - Carla Fernández Prendes
- Analysis and Clinical Biochemistry Service, Laboratori Clínic Metropolitana Nord, Hospital Universitari Germans Trias i Pujol, Badalona, Barcelona, Spain
| | - Núria Amigó Grau
- Department of Basic Medical Sciences, Rovira i Virgili University, Reus, Spain
- Center for Biomedical Research in Diabetes and Associated Metabolic Diseases (CIBERDEM), Instituto de Salud Carlos III, Madrid, Comunidad de Madrid, Spain
- Biosfer Teslab, Reus, Spain
| | - Pilar Calmarza
- Service of Clinical Biochemistry, Miguel Servet University Hospital, Zaragoza, Aragón, Spain
- Centre for Networked Research in Cardiovascular Diseases (CIBERCV), Instituto de Investigacion Sanitaria Aragon, Zaragoza, Spain
| | - Silvia Camós Anguila
- Service of Clinical Biochemistry- Laboratori Clínic Girona, Hospital Universitari de Girona Doctor Josep Trueta, Catalunya, Spain
| | - Beatriz Candas Estébanez
- Clinical Laboratory, Hospital of Barcelona, Barcelona, Catalunya, Spain
- Faculty of Medicine, UVic-UCC, Vic, Spain
- Faculty of Sciences, UVic-UCC, Vic, Spain
| | - María José Castro Castro
- Biochemistry Core, Laboratori Clínic Territorial Metropolitana Sud, Bellvitge University Hospital, L’Hospitalet de Llobregat, Spain
| | - David Ceacero
- Biochemistry Core, Laboratori Clínic Territorial Metropolitana Sud, Bellvitge University Hospital, L’Hospitalet de Llobregat, Spain
| | - Irene González Martínez
- Service of Clinical Biochemistry, 12 de Octubre University Hospital, Madrid, Autonomous Community of Madrid, Spain
| | - María Martín Palencia
- Service of Clinical Biochemistry, University Hospital of Burgos, Burgos, Castilla y León, Spain
| | - José Puzo Foncillas
- Service of Clinical Biochemistry, Unit of Lipids, Hospital General Universitario San Jorge de Huesca, General University Hospital, Huesca, Spain
- Faculty of Life Sciences and Sports, Huesca, Spain
| | - Carlos Romero Román
- General University Hospital of Albacete, Albacete, Castilla-La Mancha, Spain
| | - on behalf of the SEQCML Working Group on Lipoproteins and Cardiovascular Diseases
- Virgen Macarena University Hospital, Seville, Spain
- Analysis and Clinical Biochemistry Service, Laboratori Clínic Metropolitana Nord, Hospital Universitari Germans Trias i Pujol, Badalona, Barcelona, Spain
- Department of Basic Medical Sciences, Rovira i Virgili University, Reus, Spain
- Center for Biomedical Research in Diabetes and Associated Metabolic Diseases (CIBERDEM), Instituto de Salud Carlos III, Madrid, Comunidad de Madrid, Spain
- Biosfer Teslab, Reus, Spain
- Service of Clinical Biochemistry, Miguel Servet University Hospital, Zaragoza, Aragón, Spain
- Centre for Networked Research in Cardiovascular Diseases (CIBERCV), Instituto de Investigacion Sanitaria Aragon, Zaragoza, Spain
- Service of Clinical Biochemistry- Laboratori Clínic Girona, Hospital Universitari de Girona Doctor Josep Trueta, Catalunya, Spain
- Clinical Laboratory, Hospital of Barcelona, Barcelona, Catalunya, Spain
- Faculty of Medicine, UVic-UCC, Vic, Spain
- Faculty of Sciences, UVic-UCC, Vic, Spain
- Biochemistry Core, Laboratori Clínic Territorial Metropolitana Sud, Bellvitge University Hospital, L’Hospitalet de Llobregat, Spain
- Service of Clinical Biochemistry, 12 de Octubre University Hospital, Madrid, Autonomous Community of Madrid, Spain
- Service of Clinical Biochemistry, University Hospital of Burgos, Burgos, Castilla y León, Spain
- Service of Clinical Biochemistry, Unit of Lipids, Hospital General Universitario San Jorge de Huesca, General University Hospital, Huesca, Spain
- Faculty of Life Sciences and Sports, Huesca, Spain
- General University Hospital of Albacete, Albacete, Castilla-La Mancha, Spain
| |
Collapse
|
6
|
Arrobas Velilla T, Fernández Prendes C, Amigó Grau N, Calmarza P, Camós Anguila S, Candas Estébanez B, Castro Castro MJ, Ceacero D, González Martínez I, Martín Palencia M, Puzo Foncillas J, Romero Román C, en nombre de la Comisión de Lipoproteínas y Enfermedades Cardiovasculares de la SEQC ML. Aspectos fundamentales en la solicitud y determinación de la lipoproteína(a) en el laboratorio clínico. ADVANCES IN LABORATORY MEDICINE 2025; 6:17-27. [PMID: 40160393 PMCID: PMC11949533 DOI: 10.1515/almed-2024-0090] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Accepted: 01/08/2025] [Indexed: 04/02/2025]
Abstract
Las enfermedades cardiovasculares continúan siendo la principal causa de muerte en España, lo que sugiere la necesidad de estudiar la presencia de nuevos factores de riesgo que puedan estar contribuyendo a aumentar el riesgo cardiovascular. La lipoproteína(a) (Lp(a)) se ha asociado con un mayor riesgo de desarrollar estenosis valvular aórtica, insuficiencia cardíaca, ictus isquémico, cardiopatía isquémica y enfermedad arterial periférica. La hiperlipoproteinemia(a) es un problema de salud generalizado. Entre el 10 % y el 30 % de la población mundial presenta valores de Lp(a) superiores a 50 mg/dL. La evidencia científica acumulada en los últimos años ha confirmado la existencia de una asociación independiente entre la concentración de Lp(a) y el riesgo de presentar un evento cardiovascular arteriosclerótico. Este hallazgo, unido al creciente desarrollo de nuevas terapias específicas para reducir la Lp(a), ha incrementado notablemente el interés por su medición. El objetivo de este documento es, en base a la evidencia actual, informar sobre a qué pacientes se debería medir la Lp(a), cuáles son los métodos de medición recomendados, las concentraciones deseables y la utilidad de su medición en la reclasificación de pacientes según su riesgo cardiovascular.
Collapse
Affiliation(s)
| | - Carla Fernández Prendes
- Servicio de Análisis y Bioquímica clínica. Laboratori Clínic Metropolitana Nord, Hospital Universitari Germans Trias i Pujol, Badalona, Barcelona, España
| | - Núria Amigó Grau
- Department of Basic Medical Sciences, Rovira i Virgili University, Reus, España
- Center for Biomedical Research in Diabetes and Associated Metabolic Diseases (CIBERDEM), Instituto de Salud Carlos III, Madrid, Comunidad de Madrid, España
- Biosfer Teslab, Reus, España
| | - Pilar Calmarza
- Servicio de Bioquímica Clínica, Hospital Universitario Miguel Servet, Zaragoza, Aragón, España
- Centro de Investigación en Red en Enfermedades Cardiovasculares (CIBERCV), Instituto de Investigacion Sanitaria Aragon, Zaragoza, España
| | - Silvia Camós Anguila
- Servicio de Análisis Clínicos – Bioquímica – Laboratori Clínic, Hospital Universitari de Girona Doctor Josep Trueta, Girona, Catalunya, España
| | - Beatriz Candas Estébanez
- Laboratorio Clínico, Hospital de Barcelona, Barcelona, Catalunya, España
- Facultat de Medicina, UVic-UCC, Vic, España
- Facultat de Ciències, UVic-UCC, Vic, España
| | - María José Castro Castro
- Core Bioquímica, Laboratori Clínic Territorial Metropolitana Sud, Hospital Universitario de Bellvitge, L’Hospitalet de Llobregat, España
| | - David Ceacero
- Core Bioquímica, Laboratori Clínic Territorial Metropolitana Sud, Hospital Universitario de Bellvitge, L’Hospitalet de Llobregat, España
| | - Irene González Martínez
- Servicio de Análisis Clínicos, Hospital Universitario 12 de Octubre, Madrid, Comunidad de Madrid, España
| | - María Martín Palencia
- Servicio de Análisis Clínicos, Hospital Universitario de Burgos, Burgos, Castilla y León, España
| | - José Puzo Foncillas
- Servicio de Análisis y Bioquímica Clínica. Unidad de Lípidos, Hospital General Universitario San Jorge de Huesca, Huesca, España
- Facultad de Ciencias de la Salud y Deporte, Huesca, España
| | - Carlos Romero Román
- Hospital General Universitario de Albacete, Albacete, Castilla-La Mancha, España
| | - en nombre de la Comisión de Lipoproteínas y Enfermedades Cardiovasculares de la SEQCML
- Hospital Universitario Virgen Macarena, Sevilla, España
- Servicio de Análisis y Bioquímica clínica. Laboratori Clínic Metropolitana Nord, Hospital Universitari Germans Trias i Pujol, Badalona, Barcelona, España
- Department of Basic Medical Sciences, Rovira i Virgili University, Reus, España
- Center for Biomedical Research in Diabetes and Associated Metabolic Diseases (CIBERDEM), Instituto de Salud Carlos III, Madrid, Comunidad de Madrid, España
- Biosfer Teslab, Reus, España
- Servicio de Bioquímica Clínica, Hospital Universitario Miguel Servet, Zaragoza, Aragón, España
- Centro de Investigación en Red en Enfermedades Cardiovasculares (CIBERCV), Instituto de Investigacion Sanitaria Aragon, Zaragoza, España
- Servicio de Análisis Clínicos – Bioquímica – Laboratori Clínic, Hospital Universitari de Girona Doctor Josep Trueta, Girona, Catalunya, España
- Laboratorio Clínico, Hospital de Barcelona, Barcelona, Catalunya, España
- Facultat de Medicina, UVic-UCC, Vic, España
- Facultat de Ciències, UVic-UCC, Vic, España
- Core Bioquímica, Laboratori Clínic Territorial Metropolitana Sud, Hospital Universitario de Bellvitge, L’Hospitalet de Llobregat, España
- Servicio de Análisis Clínicos, Hospital Universitario 12 de Octubre, Madrid, Comunidad de Madrid, España
- Servicio de Análisis Clínicos, Hospital Universitario de Burgos, Burgos, Castilla y León, España
- Servicio de Análisis y Bioquímica Clínica. Unidad de Lípidos, Hospital General Universitario San Jorge de Huesca, Huesca, España
- Facultad de Ciencias de la Salud y Deporte, Huesca, España
- Hospital General Universitario de Albacete, Albacete, Castilla-La Mancha, España
| |
Collapse
|
7
|
Lai Y, Zhang S, Guo Y, Xu C, Huang M, Zhan R, Liu M, Xiong Z, Huang Y, Liao X, Ma Y, Zhuang X, Jiang J, Wu X. Apolipoprotein B modifies the association between lipoprotein(a) and ASCVD risk. Am Heart J 2025; 281:157-167. [PMID: 39643097 DOI: 10.1016/j.ahj.2024.11.014] [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: 05/14/2024] [Revised: 11/25/2024] [Accepted: 11/28/2024] [Indexed: 12/09/2024]
Abstract
BACKGROUND Elevated lipoprotein(a) (Lp[a]) and apolipoprotein B (apoB) are individually associated with the risk of atherosclerotic cardiovascular disease (ASCVD). Moreover, previous basic research has implicated the potential interaction between apoB and Lp(a) in the atherogenic process. We aimed to determine whether apoB levels significantly modulate ASCVD risk associated with Lp(a) in a large community-based population without baseline cardiovascular disease. METHODS Plasma Lp(a) and apoB were measured in the Atherosclerosis Risk in Communities (ARIC) study. Elevated Lp(a) was defined as the highest race-specific quintile, and elevated apoB was defined as ≥89 mg/dl (median value). The modifying effect of apoB on the Lp(a)-related risk of ASCVD and coronary heart disease (CHD) was determined using Cox regression models adjusted for cardiovascular risk factors. RESULTS Among 12,988 ARIC participants, 3,888 ASCVD events and 1754 CHD events were observed. Elevated apoB (≥89 mg/dl) and elevated Lp(a) (race-specific quintile 5) were independently associated with ASCVD (hazard ratio [HR]: 1.19; 95% CI: 1.08-1.30; P <0.001; HR: 1.27; 95% CI: 1.16-1.40; P < .001, respectively). Lp(a)-by-apoB interaction was noted [Lp(a) (quintile 1-4 or quintile 5) * apoB (<89 or ≥89 mg/dl) = 0.002]. Compared to the concordantly low Lp(a) group, the individuals with high Lp(a) had a greater ASCVD risk only when apoB was elevated (HR: 1.48; 95% CI: 1.34-1.63; P < .001). CONCLUSIONS In the context of primary prevention, ASCVD risk associated with Lp(a) was observed only when apoB was elevated. The measurement of apoB can further refine and contextualize the ASCVD risk associated with Lp(a).
Collapse
Affiliation(s)
- Yuhui Lai
- Cardiology Department, First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China; NHC Key Laboratory of Assisted Circulation, Sun Yat-sen University, Guangzhou, China
| | - Shaozhao Zhang
- Cardiology Department, First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China; NHC Key Laboratory of Assisted Circulation, Sun Yat-sen University, Guangzhou, China
| | - Yue Guo
- Cardiology Department, First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China; NHC Key Laboratory of Assisted Circulation, Sun Yat-sen University, Guangzhou, China
| | - Chaoguang Xu
- Cardiology Department, First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China; NHC Key Laboratory of Assisted Circulation, Sun Yat-sen University, Guangzhou, China
| | - Mengting Huang
- Cardiology Department, First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China; NHC Key Laboratory of Assisted Circulation, Sun Yat-sen University, Guangzhou, China
| | - Rongjian Zhan
- Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Menghui Liu
- Cardiology Department, First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China; NHC Key Laboratory of Assisted Circulation, Sun Yat-sen University, Guangzhou, China
| | - Zhenyu Xiong
- Cardiology Department, First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China; NHC Key Laboratory of Assisted Circulation, Sun Yat-sen University, Guangzhou, China
| | - Yiquan Huang
- Cardiology Department, First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China; NHC Key Laboratory of Assisted Circulation, Sun Yat-sen University, Guangzhou, China
| | - Xinxue Liao
- Cardiology Department, First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China; NHC Key Laboratory of Assisted Circulation, Sun Yat-sen University, Guangzhou, China
| | - Yuedong Ma
- Cardiology Department, First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China; NHC Key Laboratory of Assisted Circulation, Sun Yat-sen University, Guangzhou, China
| | - Xiaodong Zhuang
- Cardiology Department, First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China; NHC Key Laboratory of Assisted Circulation, Sun Yat-sen University, Guangzhou, China
| | - Jingzhou Jiang
- Cardiology Department, First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China.
| | - Xing Wu
- Cardiology Department, First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China.
| |
Collapse
|
8
|
Pavanello C, Ruscica M, Castiglione S, Mombelli GG, Alberti A, Calabresi L, Sirtori CR. Triglyceride-glucose index: carotid intima-media thickness and cardiovascular risk in a European population. Cardiovasc Diabetol 2025; 24:17. [PMID: 39806381 PMCID: PMC11731386 DOI: 10.1186/s12933-025-02574-2] [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: 10/22/2024] [Accepted: 01/02/2025] [Indexed: 01/16/2025] Open
Abstract
BACKGROUND The triglyceride-glucose (TyG) index is now widely recognized as a marker of insulin resistance and has been linked to the development and prognosis of atherosclerotic cardiovascular diseases (ASCVD) in numerous populations, particularly in the Eastern world. Although there are fewer reports from the Western world, and they are sometimes contradictory, the absence of definitive data on the relationship between a raised TyG index and cardiovascular risk suggested the opportunity of testing this biochemical marker against a well-established vascular marker such as the carotid intima media thickness (c-IMT). METHODS Primary prevention patients were selected from a cohort of individuals who underwent c-IMT measurement between 1984 and 2018 at the Dyslipidemia Center at the ASST Grande Ospedale Metropolitano Niguarda in Milan, Italy. The TyG index was calculated as the Ln [fasting TG (mg/dL)×fasting glucose (mg/dL)/2]. Carotid ultrasonography was performed using echographic measurements of the far walls of the left and right common, internal carotids, and bifurcations. Patients were followed for up to 20 years with periodic evaluation of biochemical parameters. ASCVD events were monitored through hospital records, where all patients were regularly examined. RESULTS The analysis included 3108 individuals with a mean age of 54.9 ± 13.1 years. Participants were generally non-obese, with an average BMI of 24.6 ± 3.5 Kg/m2. Among the women, 83.1% were postmenopausal. The mean TyG index was 8.65 ± 0.59. There was a significant association between the TyG index and all c-IMT measurements. Those in the highest TyG index quartiles had significantly higher IMTmean and IMTmax compared to those in the lower quartiles. These associations were consistent across all vascular sites examined and remained significant after adjusting for all potential confounders. Kaplan-Meier survival analysis revealed an increased incidence of ASCVD events in the two highest TyG index quartiles. CONCLUSIONS TyG index is a sensitive marker of risk in a European population with moderate ASCVD risk, as assessed by c-IMT measurements, in a large cohort of Lipid Clinic patients.
Collapse
Affiliation(s)
- Chiara Pavanello
- Centro E. Grossi Paoletti, Dipartimento di Scienze Farmacologiche e Biomolecolari "Rodolfo Paoletti", Università degli Studi di Milano, Milano, Italy.
- Dyslipidemia Center, SSD Diagnosi e Cure Territoriali Malattie Cardiache, ASST Grande Ospedale Metropolitano Niguarda, Milan, Italy.
| | - Massimiliano Ruscica
- Dipartimento di Scienze Farmacologiche e Biomolecolari "Rodolfo Paoletti", Università degli Studi di Milano, Milan, Italy
- Department of Cardio-Thoracic-Vascular Diseases, Foundation IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Sofia Castiglione
- Centro E. Grossi Paoletti, Dipartimento di Scienze Farmacologiche e Biomolecolari "Rodolfo Paoletti", Università degli Studi di Milano, Milano, Italy
- Dyslipidemia Center, SSD Diagnosi e Cure Territoriali Malattie Cardiache, ASST Grande Ospedale Metropolitano Niguarda, Milan, Italy
| | - Giuliana Germana Mombelli
- Dyslipidemia Center, SSD Diagnosi e Cure Territoriali Malattie Cardiache, ASST Grande Ospedale Metropolitano Niguarda, Milan, Italy
| | - Antonia Alberti
- Dyslipidemia Center, SSD Diagnosi e Cure Territoriali Malattie Cardiache, ASST Grande Ospedale Metropolitano Niguarda, Milan, Italy
| | - Laura Calabresi
- Centro E. Grossi Paoletti, Dipartimento di Scienze Farmacologiche e Biomolecolari "Rodolfo Paoletti", Università degli Studi di Milano, Milano, Italy
| | - Cesare Riccardo Sirtori
- Centro E. Grossi Paoletti, Dipartimento di Scienze Farmacologiche e Biomolecolari "Rodolfo Paoletti", Università degli Studi di Milano, Milano, Italy.
- Dipartimento di Scienze Farmacologiche e Biomolecolari "Rodolfo Paoletti", Università degli Studi di Milano, Milan, Italy.
| |
Collapse
|
9
|
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
|
10
|
Liu J, Liu Z, Xu D, Zhou T, Li A, Hu J, Li H, Li W, Wang Z, Yu Z, Zeng L. Pretreatment Lipoprotein(a) as a Biomarker for EGFR Mutation and Prognosis in Lung Adenocarcinoma. Int J Gen Med 2024; 17:6465-6478. [PMID: 39742033 PMCID: PMC11687294 DOI: 10.2147/ijgm.s501401] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2024] [Accepted: 12/11/2024] [Indexed: 01/03/2025] Open
Abstract
Purpose This study aims to investigate the correlation between pretreatment serum lipoprotein(a) [Lp(a)] and epidermal growth factor receptor (EGFR) gene mutations, as well as its predictive value for progression-free survival (PFS) in advanced lung adenocarcinoma patients receiving epidermal growth factor receptor-tyrosine kinase inhibitors (EGFR-TKIs) therapy. Patients and Methods We determined the optimal cutoff value for Lp(a) by receiver operating characteristic (ROC) curves and Youden's index to categorize Lp(a) into high and low groups. Logistic regression was used to analyze the EGFR mutation rate in different groups. Additionally, the relationship between pretreatment Lp(a) levels and prognostic PFS in patients with advanced (TNM stage IIIB-IV) lung adenocarcinoma treated with EGFR-TKIs was retrospectively analyzed by Cox regression, survival and stratified analysis methods. Results We included 338 advanced lung adenocarcinoma patients, with median age of 64 years, and slightly more female patients (51.8%), most of whom had no smoking history (70.7%), no history of chronic lung disease (87.9%), and stage IV (81.1%) patients. The EGFR gene mutation rate was 55.3% and 123 patients were included in the prognostic evaluation through screening. The optimal cutoff value for Lp(a) was 20.48 mg/L. The mutation rate in the high Lp(a) group was significantly lower than the low Lp(a) group (48.0% vs 65.5%, p = 0.001). Multivariate logistic regression analysis indicated that Lp(a) is an independent predictor of EGFR mutations (OR = 0.41, 95% CI: 0.25-0.66, p<0.001). Survival analysis showed that the median PFS was significantly longer in the high Lp(a) level group compared to the low level group (16.1 months, 95% CI: 11.9-23.8 months vs 9.6 months, 95% CI: 8.9-13.3 months, p=0.015). Multivariate analysis confirmed that Lp(a) is an independent predictor of PFS in advanced lung adenocarcinoma patients receiving EGFR-TKIs treatment (HR = 0.42, 95% CI: 0.26-0.68, p<0.001). Conclusion Pretreatment Lp(a) may be a biomarker for EGFR mutations and the PFS in advanced lung adenocarcinoma patients undergoing EGFR-TKIs treatment.
Collapse
Affiliation(s)
- Ji Liu
- Department of Respiratory and Critical Care Medicine, the Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province, People’s Republic of China
| | - Zhekang Liu
- Rheumatology and Immunology department, the Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province, People’s Republic of China
| | - Deming Xu
- Department of Respiratory and Critical Care Medicine, the Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province, People’s Republic of China
| | - Tao Zhou
- Department of Respiratory and Critical Care Medicine, the Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province, People’s Republic of China
| | - Ang Li
- Department of Respiratory and Critical Care Medicine, the Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province, People’s Republic of China
| | - Jiali Hu
- Department of Respiratory and Critical Care Medicine, the Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province, People’s Republic of China
| | - Hong Li
- Department of Geriatrics, the First People’s Hospital of Jiashan County, Jiaxing, Zhejiang Province, People’s Republic of China
| | - Wenjie Li
- Department of Respiratory and Critical Care Medicine, the Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province, People’s Republic of China
| | - Zengqing Wang
- Department of Respiratory and Critical Care Medicine, the Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province, People’s Republic of China
| | - Zhiping Yu
- Department of Respiratory and Critical Care Medicine, the Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province, People’s Republic of China
| | - Linxiang Zeng
- Department of Respiratory and Critical Care Medicine, the Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province, People’s Republic of China
| |
Collapse
|
11
|
Fan H, Kouvari M, Mingrone G, George J, Papatheodoridis G, Valenzuela-Vallejo L, Liu Z, Chen X, Zhang T, Mantzoros CS. Lipoprotein (a) in the Full Spectrum of Metabolic Dysfunction-associated Steatotic Liver Disease: Evidence From Histologically and Genetically Characterized Cohorts. Clin Gastroenterol Hepatol 2024:S1542-3565(24)01065-6. [PMID: 39672249 DOI: 10.1016/j.cgh.2024.10.018] [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] [Received: 08/21/2024] [Revised: 10/02/2024] [Accepted: 10/11/2024] [Indexed: 12/15/2024]
Abstract
BACKGROUND & AIMS Lipoprotein(a) (Lp(a)) is an emerging biomarker for cardiometabolic factors. We studied the role of Lp(a) in the full spectrum of metabolic dysfunction-associated steatotic liver disease (MASLD). METHODS Three independent analyses were implemented using a multi-center, cross-sectional, liver biopsy-based study (n = 332) (Study 1) and the UK Biobank prospective study (n = 270,004) (Study 2; median follow-up, 12.47 years). In Study 1, we studied the cross-sectional association between Lp(a) mass and MASLD stages (Analysis A). In Study 2, we studied the prospective association between Lp(a) concentration and MASLD, liver cirrhosis, and hepatocellular carcinoma (Analysis B). Finally, these analyses were accompanied by a prospective analysis using LPA Genetic Risk Score (Analysis C). RESULTS In Study 1, an inverse association between Lp(a) and at-risk metabolic dysfunction-associated steatohepatitis (odds ratioper 1-SD increase, 0.64; 95% confidence interval [CI], 0.42-0.97) was observed. In contrast, when similar associations were examined prospectively (Study 2), subjects with Lp(a) <10.75 nmol/L had a higher risk for cirrhosis (hazard ratio, 1.49; 95% CI, 1.22-1.81) and HCC (hazard ratio, 1.69; 95% CI, 1.12-2.56) compared with subjects with Lp(a) within the 10.75 to 21.5 nmol/L range. Above these levels, the risk increased significantly and positively. Similarly, genetic analysis showed an L-shaped association with LPA Genetic Risk Score. CONCLUSIONS The inverse association observed in cross-sectional studies should be attributed to reverse causality (ie, the presence of liver disease may decrease Lp(a) levels). Genetically predicted very low Lp(a) levels are also associated with impaired liver health prospectively. Clinical trials evaluating Lp(a)-lowering agents should thus be monitored carefully for adverse liver effects in subjects attaining extremely low concentrations.
Collapse
Affiliation(s)
- Hong Fan
- Department of Epidemiology, School of Public Health, Fudan University, Shanghai, China; Fudan University Taizhou Institute of Health Sciences, Taizhou, China; Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
| | - Matina Kouvari
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts.
| | | | - Jacob George
- Storr Liver Centre, Westmead Institute for Medical Research, Westmead Hospital, Westmead, New South Wales, Australia; University of Sydney, Sydney, New South Wales, Australia
| | - Georgios Papatheodoridis
- Department of Gastroenterology, Medical School of National and Kapodistrian University of Athens, General Hospital of Athens "Laiko," Athens, Greece
| | - Laura Valenzuela-Vallejo
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
| | - Zhenqiu Liu
- Fudan University Taizhou Institute of Health Sciences, Taizhou, China; State Key Laboratory of Genetic Engineering, Human Phenome Institute, Fudan University, Shanghai, China; School of Life Sciences, Fudan University, Shanghai, China
| | - Xingdong Chen
- Fudan University Taizhou Institute of Health Sciences, Taizhou, China; State Key Laboratory of Genetic Engineering, Human Phenome Institute, Fudan University, Shanghai, China; School of Life Sciences, Fudan University, Shanghai, China
| | - Tiejun Zhang
- Department of Epidemiology, School of Public Health, Fudan University, Shanghai, China; Fudan University Taizhou Institute of Health Sciences, Taizhou, China.
| | - Christos S Mantzoros
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts; Section of Endocrinology, VA Boston Healthcare System, Jamaica Plain, Massachusetts.
| |
Collapse
|
12
|
Fichtner I, Macchi C, Rizzuto AS, Carugo S, Corsini A, Ruscica M. Lipoprotein(a) and the atherosclerotic burden - Should we wait for clinical trial evidence before taking action? ATHEROSCLEROSIS PLUS 2024; 58:16-23. [PMID: 39435317 PMCID: PMC11492331 DOI: 10.1016/j.athplu.2024.09.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/24/2024] [Revised: 09/20/2024] [Accepted: 09/22/2024] [Indexed: 10/23/2024]
Abstract
The fact that lipoprotein(a) levels should be regarded as a causal residual risk factor in the atherosclerotic cardiovascular diseases (ASCVD) is now a no-brainer. This review article aims to summarize the latest evidence supporting the causal role of lipoprotein(a) in ASCVD and the potential strategies to reduce the lipoprotein(a) burden until clinical trial results are available. Epidemiological and genetic data demonstrate the causal link between lipoprotein(a) and increased ASCVD risk. That being said, a specific question comes to mind: "must we wait for outcome trials in order to take action?". Given that lipoprotein(a) levels predict incident ASCVD in both primary and secondary prevention contexts, with a linear risk gradient across its distribution, measuring lipoprotein(a) can unequivocally help identify patients who may later benefit from specific lipoprotein(a)-lowering therapies. This understanding has led various National Societies to recommend dosing lipoprotein(a) in high-risk individuals and to support the recommendation of measuring lipoprotein(a) levels at least once in every adult for risk stratification.
Collapse
Affiliation(s)
- Isabella Fichtner
- Department of Pharmacological and Biomolecular Sciences "Rodolfo Paoletti", Università Degli Studi Di Milano, Milan, Italy
| | - Chiara Macchi
- Department of Pharmacological and Biomolecular Sciences "Rodolfo Paoletti", Università Degli Studi Di Milano, Milan, Italy
| | | | - Stefano Carugo
- Department of Clinical Sciences and Community Health, Università Degli Studi Di Milano, Milan, Italy
- Department of Cardio-Thoracic-Vascular Diseases, Foundation IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Alberto Corsini
- Department of Pharmacological and Biomolecular Sciences "Rodolfo Paoletti", Università Degli Studi Di Milano, Milan, Italy
| | - Massimiliano Ruscica
- Department of Pharmacological and Biomolecular Sciences "Rodolfo Paoletti", Università Degli Studi Di Milano, Milan, Italy
- Department of Cardio-Thoracic-Vascular Diseases, Foundation IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| |
Collapse
|
13
|
Zhang N, Shi K, Ma S, Zhang X, Duan M, Zhang M, Liu Y, Gao T, Yang H, Ma X, Huang Y, Cheng Y, Qu H, Fan J, Yao Q, Zhan S. Correlation between lipoprotein(a) and recurrent ischemic events post-cerebral vascular stent implantation. J Stroke Cerebrovasc Dis 2024; 33:107882. [PMID: 39038628 DOI: 10.1016/j.jstrokecerebrovasdis.2024.107882] [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: 05/14/2024] [Revised: 07/16/2024] [Accepted: 07/19/2024] [Indexed: 07/24/2024] Open
Abstract
BACKGROUND AND AIM The association of Lipoprotein(a) (Lp[a]) with recurrent ischemic events in stented patients remains uncertain. So, this research aimed to investigate the impact of elevated Lp(a) levels on the occurrence of ischemic events in this specific patient population. METHODS Totally 553 patients who underwent intracranial or extracranial artery stent implantation were included. Baseline data were collected and postoperative ischemic outcomes were followed up. Cox regression analysis was used to investigate the association between Lp(a) and outcomes, while accounting for confounding factors. Finally, we established prediction models based on nomogram. RESULTS Of total 553 patients, a number of 107 (19.3%) experienced outcomes. These included 46 cases (25.4%) in group with elevated Lp(a) levels (>30 mg/dL) and 61 cases (16.4%) in non-elevated group (χ2=6.343, p=0.012). The group with elevated Lp(a) was 1.811 times more likely to experience ischemic events than the non-elevated group, each 1 mg/dL increase in Lp(a) resulted in a 1.008-fold increase in the recurrence rate of ischemic events. In addition, sex (male), previous history of coronary heart disease, decreased albumin, elevated very low density lipoprotein cholesterol and poorly controlled risk factors (including blood pressure and blood sugar) were also associated with a high risk of recurrent ischemic events after stent implantation. CONCLUSION Lp(a) elevation was a significant risk factor for ischemic events in symptomatic patients who underwent intracranial or extracranial artery stenting.
Collapse
Affiliation(s)
- Nan Zhang
- Department of Neurology, the Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710004, China
| | - Kaili Shi
- Department of Neurology, the Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710004, China
| | - Shuyin Ma
- Department of Neurology, the Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710004, China
| | - Xiaodong Zhang
- Department of Neurology, the Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710004, China
| | - Minyu Duan
- Department of Neurology, the Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710004, China
| | - Mengyuan Zhang
- Department of Neurology, the Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710004, China
| | - Yixin Liu
- Department of Neurology, the Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710004, China
| | - Tiantian Gao
- Department of Neurology, the Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710004, China
| | - Han Yang
- Department of Neurology, the Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710004, China
| | - Xiaodong Ma
- Department of Neurology, the Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710004, China
| | - Yizhou Huang
- Department of Neurology, the Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710004, China
| | - Yuxuan Cheng
- Department of Neurology, the Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710004, China
| | - Huiyang Qu
- Department of Neurology, the Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710004, China
| | - Jiaxin Fan
- Department of Neurology, the Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710004, China
| | - Qingling Yao
- Department of Neurology, the Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710004, China
| | - Shuqin Zhan
- Department of Neurology, the Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710004, China.
| |
Collapse
|
14
|
Kamel AK, Farag NM, Allam E, Khaled M, Ismail DE. Expression of Monocytes Subsets in Patients Diagnosed With Coronary Artery Atherosclerosis and Their Impact on Disease Severity. Cureus 2024; 16:e74670. [PMID: 39734983 PMCID: PMC11681966 DOI: 10.7759/cureus.74670] [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] [Accepted: 11/27/2024] [Indexed: 12/31/2024] Open
Abstract
Introduction Many studies have supported inflammation as a mediator of lipoprotein (a) (Lp(a)) induced increase in cardiovascular disease risk, as it has pro-inflammatory effects on endothelial cells and monocytes. Aim This study aims to correlate Lp(a) level with different monocyte subsets in coronary atherosclerotic patients with different severity. Method The study included 60 patients with a mean age of 53.1 ± 10.5 diagnosed as coronary atherosclerotic patients by coronary angiography. Lp levels were measured using enzyme-linked immunosorbent assay (ELISA), while blood counts and monocyte subsets were analyzed by flow cytometry, and 30 apparently healthy individuals were included as the control group. Results Patients showed significantly higher median monocytic %, Lp(a), and higher C-reactive protein (CRP) values than the control group. Patients were subdivided into two groups: normal Lp(a) < 6.2 mg/dL (n = 24) and hyperlipoproteinemia(a) (hyper Lp(a)) ≥ 6.2 mg/dL (n = 36). Patients with hyper Lp(a) had higher non-classical monocytes (31.5% vs. 20%). Coronary atherosclerosis severity was associated with higher Lp(a) levels as well as non-classical monocytes; patients with mild atherosclerosis showed the highest classical and intermediate subset levels. While for a non-classical subset, patients with severe atherosclerosis showed the highest median level. A significant moderate positive correlation between Lp(a) and monocyte counts, as well as monocyte-lymphocyte (M/L) index and non-classical monocytes, was found. Conclusions Hyper Lp(a) and increased count of non-classical monocytes are significantly increased with disease progression (triple-vessel coronary disease risk). These results suggest that the expansion of non-classical monocytes is a cardiovascular disease (CVD) risk and predictor for disease severity. Strategies targeting inflammatory monocytes may slow CVD progression.
Collapse
Affiliation(s)
- Asmaa Khalf Kamel
- Department of Clinical Pathology, Minia University Faculty of Medicine, Minia, EGY
| | - Naglaa M Farag
- Department of Clinical Pathology, Minia University Faculty of Medicine, Minia, EGY
| | - Emad Allam
- Department of Clinical Pathology, Minia University Faculty of Medicine, Minia, EGY
| | - Mohamed Khaled
- Department of Cardiology, Minia University Faculty of Medicine, Minia, EGY
| | - Doaa Elzaeem Ismail
- Department of Clinical Pathology, Minia University Faculty of Medicine, Minia, EGY
| |
Collapse
|
15
|
Li S, Liu H, Zhang Y, Zhang M, Zhang H, Zhu C, Wu N, Xu R, Dong Q, Qian J, Dou K, Guo Y, Li J. Prognostic role of lipoprotein(a) in atherosclerotic cardiovascular disease risk from a perspective on current risk stratification. MedComm (Beijing) 2024; 5:e773. [PMID: 39492837 PMCID: PMC11527816 DOI: 10.1002/mco2.773] [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: 12/24/2023] [Revised: 08/14/2024] [Accepted: 09/04/2024] [Indexed: 11/05/2024] Open
Abstract
Lipoprotein(a) [Lp(a)] is an emerging predictor for atherosclerotic cardiovascular disease (ASCVD) but the association from a perspective on current risk stratification was unknown. A cohort of 9944 Chinese patients with ASCVD was recruited and refined into very-high-risk (VHR) and non-VHR subgroups according to current guideline. Lp(a) plasma levels were divided by its concentration (<30, 30-50, 50-75, and ≥75 mg/dL) and percentile zones (<25th, 25-50th, 50-75th, 75-90th, ≥90th). Cardiovascular events (CVEs) occurred during an average of 38.5 months' follow-up were recorded. We found that Lp(a) was increased with risk stratification of ASCVD increasing. Prevalence of CVEs had a significantly increasing trend with gradients of Lp(a) elevation in VHR but not in non-VHR subgroup. The adjusted HRs (95%CIs) for CVEs were 1.75(1.25-2.46) in the highest group of Lp(a) ≥75 mg/dL compared with the group of Lp(a) <30 mg/dL as the reference in overall patients, 2.18(1.32-3.58) in VHR subgroup and 1.43(0.93-2.18) in non-VHR subgroup, respectively. The adjusted HRs (95%CIs) at the highest grade of Lp(a) levels (≥90th) were 1.72(1.19-2.50) in overall population, 2.83(1.53-5.24) in VHR subgroup and 1.38(0.86-2.12) in non-VHR subgroup, respectively. These findings suggested that Lp(a) might contribute more to CVEs risk in VHR subgroup of ASCVD.
Collapse
Affiliation(s)
- Sha Li
- Cardiometabolic CenterState Key Laboratory of Cardiovascular DiseaseFuWai HospitalNational Center for Cardiovascular DiseasesChinese Academy of Medical SciencesPeking Union Medical CollegeBeijingChina
| | - Hui‐Hui Liu
- Cardiometabolic CenterState Key Laboratory of Cardiovascular DiseaseFuWai HospitalNational Center for Cardiovascular DiseasesChinese Academy of Medical SciencesPeking Union Medical CollegeBeijingChina
| | - Yan Zhang
- Cardiometabolic CenterState Key Laboratory of Cardiovascular DiseaseFuWai HospitalNational Center for Cardiovascular DiseasesChinese Academy of Medical SciencesPeking Union Medical CollegeBeijingChina
| | - Meng Zhang
- Cardiometabolic CenterState Key Laboratory of Cardiovascular DiseaseFuWai HospitalNational Center for Cardiovascular DiseasesChinese Academy of Medical SciencesPeking Union Medical CollegeBeijingChina
| | - Hui‐Wen Zhang
- Cardiometabolic CenterState Key Laboratory of Cardiovascular DiseaseFuWai HospitalNational Center for Cardiovascular DiseasesChinese Academy of Medical SciencesPeking Union Medical CollegeBeijingChina
| | - Cheng‐Gang Zhu
- Cardiometabolic CenterState Key Laboratory of Cardiovascular DiseaseFuWai HospitalNational Center for Cardiovascular DiseasesChinese Academy of Medical SciencesPeking Union Medical CollegeBeijingChina
| | - Na‐Qiong Wu
- Cardiometabolic CenterState Key Laboratory of Cardiovascular DiseaseFuWai HospitalNational Center for Cardiovascular DiseasesChinese Academy of Medical SciencesPeking Union Medical CollegeBeijingChina
| | - Rui‐Xia Xu
- Cardiometabolic CenterState Key Laboratory of Cardiovascular DiseaseFuWai HospitalNational Center for Cardiovascular DiseasesChinese Academy of Medical SciencesPeking Union Medical CollegeBeijingChina
| | - Qian Dong
- Cardiometabolic CenterState Key Laboratory of Cardiovascular DiseaseFuWai HospitalNational Center for Cardiovascular DiseasesChinese Academy of Medical SciencesPeking Union Medical CollegeBeijingChina
| | - Jie Qian
- Cardiometabolic CenterState Key Laboratory of Cardiovascular DiseaseFuWai HospitalNational Center for Cardiovascular DiseasesChinese Academy of Medical SciencesPeking Union Medical CollegeBeijingChina
| | - Ke‐Fei Dou
- Cardiometabolic CenterState Key Laboratory of Cardiovascular DiseaseFuWai HospitalNational Center for Cardiovascular DiseasesChinese Academy of Medical SciencesPeking Union Medical CollegeBeijingChina
| | - Yuan‐Lin Guo
- Cardiometabolic CenterState Key Laboratory of Cardiovascular DiseaseFuWai HospitalNational Center for Cardiovascular DiseasesChinese Academy of Medical SciencesPeking Union Medical CollegeBeijingChina
| | - Jian‐Jun Li
- Cardiometabolic CenterState Key Laboratory of Cardiovascular DiseaseFuWai HospitalNational Center for Cardiovascular DiseasesChinese Academy of Medical SciencesPeking Union Medical CollegeBeijingChina
| |
Collapse
|
16
|
Lin CY, Huey-Jen Hsu S, Lee HL, Wang C, Sung FC, Su TC. Examining a decade-long trend in exposure to per- and polyfluoroalkyl substances and their correlation with lipid profiles: Insights from a prospective cohort study on the young Taiwanese population. CHEMOSPHERE 2024; 364:143072. [PMID: 39128777 DOI: 10.1016/j.chemosphere.2024.143072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/02/2024] [Revised: 07/07/2024] [Accepted: 08/08/2024] [Indexed: 08/13/2024]
Abstract
Per- and polyfluoroalkyl substances (PFAS) are artificial chemicals extensively utilized in everyday products, and numerous cross-sectional epidemiological studies consistently link PFAS exposure with lipid profiles across diverse populations and age groups. In longitudinal studies, the findings also indicate a positive correlation between PFAS and lipid profiles; however, this association remains unexplored in adolescents and young adults. Notably, previous research has predominantly focused on conventional lipid biomarkers, with limited exploration of the relationship between PFAS and diverse lipoprotein subfractions. Furthermore, there is a lack of comprehensive investigation into the temporal trends in PFAS concentrations in Taiwan. To address this research gap, we conducted a prospective study following 592 adolescents and young adults (12-30 years old at enrollment) from the YOung TAiwanese Cohort (YOTA) over a duration of 10 years. During the follow-up period, we measured 11 types of PFAS and various lipid profile biomarkers (low-density lipoprotein cholesterol (LDL-C), small dense LDL-C (sdLDL-C), low-density lipoprotein triglyceride (LDL-TG), high-density lipoprotein cholesterol (HDL-C), HDL3-C, lipoprotein(a), triglyceride). Our results revealed a general decline in PFAS concentrations in the study population. Regarding the correlation between the average levels (averaged across the initial and second tracking periods) of PFAS and lipid profiles (during the second tracking period), we observed positive correlations with total cholesterol and LDL-C for perfluorononanoic acid (PFNA), perfluoroundecanoic acid (PFUdA), perfluorododecanoic acid (PFDoA), N-methylperfluorooctane sulfonamide acetic acid (N-MeFOSAA), and the sum of PFAS (sum of the 11 kinds of PFAS). Additionally, average levels of PFUdA, N-MeFOSAA, and the sum of PFAS exhibited positive associations with sdLDL-C. This study unveiled an overall decrease in PFAS concentrations and underscores a potential link between PFAS exposure and adverse changes in lipid profiles among young populations, emphasizing the need for further exploration into the mechanisms of PFAS on lipid metabolism and atherosclerosis.
Collapse
Affiliation(s)
- Chien-Yu Lin
- Department of Internal Medicine, En Chu Kong Hospital, New Taipei City, 237, Taiwan; School of Medicine, Fu Jen Catholic University, New Taipei City, 242, Taiwan; Department of Environmental Engineering and Health, Yuanpei University of Medical Technology, Hsinchu, 300, Taiwan
| | - Sandy Huey-Jen Hsu
- Department of Laboratory Medicine, College of Medicine, National Taiwan University Hospital, National Taiwan University, Taipei, 100, Taiwan
| | - Hui-Ling Lee
- Department of Chemistry, Fu Jen Catholic University, New Taipei City, 242, Taiwan
| | - Chikang Wang
- Department of Environmental Engineering and Health, Yuanpei University of Medical Technology, Hsinchu, 300, Taiwan
| | - Fung-Chang Sung
- Department of Health Services Administration, China Medical University College of Public Health, Taichung, 404, Taiwan; Department of Food Nutrition and Health Biotechnology, Asia University, Taichung, 413, Taiwan
| | - Ta-Chen Su
- Department of Environmental and Occupational Medicine, National Taiwan University Hospital, Taipei, 100, Taiwan; Division of Cardiology, Department of Internal Medicine, National Taiwan University Hospital, Taipei, 100, Taiwan; Institute of Environmental and Occupational Health Sciences, College of Public Health, National Taiwan University, Taipei, 100, Taiwan; School of Medicine, College of Medicine, National Cheng Kung University, Tainan, 701, Taiwan.
| |
Collapse
|
17
|
Lejawa M, Goławski M, Fronczek M, Osadnik T, Paneni F, Ruscica M, Pawlas N, Lisik M, Banach M. Causal associations between insulin and Lp(a) levels in Caucasian population: a Mendelian randomization study. Cardiovasc Diabetol 2024; 23:316. [PMID: 39210428 PMCID: PMC11360791 DOI: 10.1186/s12933-024-02389-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/16/2024] [Accepted: 08/01/2024] [Indexed: 09/04/2024] Open
Abstract
BACKGROUND Numerous observational studies have demonstrated that circulating lipoprotein(a) [Lp(a)] might be inversely related to the risk of type 2 diabetes (T2D). However, recent Mendelian randomization (MR) studies do not consistently support this association. The results of in vitro research suggest that high insulin concentrations can suppress Lp(a) levels by affecting apolipoprotein(a) [apo(a)] synthesis. This study aimed to identify the relationship between genetically predicted insulin concentrations and Lp(a) levels, which may partly explain the associations between low Lp(a) levels and increased risk of T2D. METHODS Independent genetic variants strongly associated with fasting insulin levels were identified from meta-analyses of genome-wide association studies in European populations (GWASs) (N = 151,013). Summary level data for Lp(a) in the population of European ancestry were acquired from a GWAS in the UK Biobank (N = 361,194). The inverse-variance weighted (IVW) method approach was applied to perform two-sample summary-level MR. Robust methods for sensitivity analysis were utilized, such as MR‒Egger, the weighted median (WME) method, MR pleiotropy residual sum and outlier (MR-PRESSO), leave-one-out analysis, and MR Steiger. RESULTS Genetically predicted fasting insulin levels were negatively associated with Lp(a) levels (β = - 0.15, SE = 0.05, P = 0.003). The sensitivity analysis revealed that WME (β = - 0.26, SE = 0.07, P = 0.0002), but not MR‒Egger (β = - 0.22, SE = 0.13, P = 0.11), supported a causal relationship between genetically predisposed insulin levels and Lp(a). CONCLUSION Our MR study provides robust evidence supporting the association between genetically predicted increased insulin concentrations and decreased concentrations of Lp(a). These findings suggest that hyperinsulinaemia, which typically accompanies T2D, can partially explain the inverse relationship between low Lp(a) concentrations and an increased risk of T2D.
Collapse
Affiliation(s)
- Mateusz Lejawa
- Department of Pharmacology, Faculty of Medical Sciences in Zabrze, Medical University of Silesia, Katowice, Poland.
| | - Marcin Goławski
- Department of Pharmacology, Faculty of Medical Sciences in Zabrze, Medical University of Silesia, Katowice, Poland
| | - Martyna Fronczek
- Department of Pharmacology, Faculty of Medical Sciences in Zabrze, Medical University of Silesia, Katowice, Poland
| | - Tadeusz Osadnik
- Department of Pharmacology, Faculty of Medical Sciences in Zabrze, Medical University of Silesia, Katowice, Poland
| | - Francesco Paneni
- Department of Cardiology, University Heart Center, University Hospital Zurich, Rämistrasse 100, 8091, Zurich, Switzerland
- Department of Cardiology, Center for Translational and Experimental Cardiology (CTEC), Zurich University Hospital, University of Zurich, Wagistrasse 12, 8952, Schlieren, Switzerland
| | - Massimiliano Ruscica
- Department of Cardio-Thoracic-Vascular Diseases, Foundation IRCCS Cà Granda Ospedale Maggiore Policlinico, Milan, Italy
- Department of Pharmacological and Biomolecular Sciences "Rodolfo Paoletti", Università degli Studi di Milano, Milan, Italy
| | - Natalia Pawlas
- Department of Pharmacology, Faculty of Medical Sciences in Zabrze, Medical University of Silesia, Katowice, Poland
| | - Małgorzata Lisik
- Outpatient Clinic, Maria Sklodowska-Curie National Research Institute of Oncology Gliwice Branch, Gliwice, Poland
| | - Maciej Banach
- Department of Preventive Cardiology and Lipidology, Medical University of Lodz, Lodz, Poland
| |
Collapse
|
18
|
Liang Q, Zhang G, Jiang L. Association between lipoprotein (a) and risk of atherosclerotic cardiovascular disease events among maintenance hemodialysis patients in Beijing, China: a single-center, retrospective study. BMC Nephrol 2024; 25:250. [PMID: 39090533 PMCID: PMC11295529 DOI: 10.1186/s12882-024-03690-z] [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: 01/22/2024] [Accepted: 07/30/2024] [Indexed: 08/04/2024] Open
Abstract
BACKGROUND Serum lipoprotein(a) [Lp(a)] is an independent risk factor for atherosclerotic cardiovascular disease (ASCVD) in the general population, its association with ASCVD incidence in Chinese maintenance hemodialysis (MHD) patients remains unclear. We aimed to evaluate the relationship between Lp(a) levels and ASCVD incidence among MHD patients in Beijing, China. METHODS This retrospective, observational cohort study included MHD patients at Beijing Tongren Hospital from January 1, 2013 to December 1, 2020, and followed until December 1,2023. The primary outcome was ASCVD occurrence. Kaplan-Meier survival analysis was used to evaluate ASCVD-free survival in MHD patients, with stratification based on Lp(a) levels. Cox regression analyses were conducted to assess the association between Lp(a) levels and the occurrence of ASCVD. RESULTS A total of 265 patients were enrolled in the study. The median follow-up period were 71 months.78 (29.4%) participants experienced ASCVD events, and 118 (47%) patients died, with 58 (49.1%) deaths attributed to ASCVD. Spearman rank correlation analyses revealed positive correlations between serum Lp(a) levels and LDL-c levels, and negative correlations with hemoglobin, triglyceride, serum iron, serum creatinine, and albumin levels. Multivariate Cox regression analysis showed that Lp(a) levels ≥ 30 mg/L, increased age, decreased serum albumin levels, and a history of diabetes mellitus were significantly associated with ASCVD incidence. CONCLUSIONS This study demonstrated an independent and positive association between serum Lp(a) levels and the risk of ASCVD in MHD patients, suggesting that serum Lp(a) could potentially serve as a clinical biomarker for estimating ASCVD risk in this population.
Collapse
Affiliation(s)
- Qiaojing Liang
- Department of Nephrology, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Guojuan Zhang
- Department of Nephrology, Beijing Tongren Hospital, Capital Medical University, Beijing, China.
| | - Liping Jiang
- Department of Nephrology, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
19
|
Dagli-Hernandez C, Ferreira GM, Freitas RCCD, Borges JB, Oliveira VFD, Gonçalves RM, Faludi AA, Marçal EDSR, Bastos GM, Bortolin RH, Hirata MH, Hirata RDC. Predicted deleterious variants in ABCA1, LPL, LPA and KIF6 are associated with statin response and adverse events in patients with familial hypercholesterolemia and disturb protein structure and stability. Pharmacogenet Genomics 2024; 34:91-104. [PMID: 38682317 DOI: 10.1097/fpc.0000000000000524] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/01/2024]
Abstract
OBJECTIVES This study explored the association of deleterious variants in pharmacodynamics (PD) genes with statin response and adverse effects in patients with familial hypercholesterolemia (FH) and analyzed their potential effects on protein structure and stability. METHODS Clinical and laboratory data were obtained from 144 adult FH patients treated with statins. A panel of 32 PD genes was analyzed by exon-targeted gene sequencing. Deleterious variants were identified using prediction algorithms and their structural effects were analyzed by molecular modeling studies. RESULTS A total of 102 variants were predicted as deleterious (83 missense, 8 stop-gain, 4 frameshift, 1 indel, 6 splicing). The variants ABCA1 rs769705621 (indel), LPA rs41267807 (p.Tyr2023Cys) and KIF6 rs20455 (p.Trp719Arg) were associated with reduced low-density lipoprotein cholesterol (LDLc) response to statins, and the LPL rs1801177 (p.Asp36Asn) with increased LDLc response (P < 0.05). LPA rs3124784 (p.Arg2016Cys) was predicted to increase statin response (P = 0.022), and ABCA1 rs769705621 to increase the risk of statin-related adverse events (SRAE) (P = 0.027). LPA p.Arg2016Cys and LPL p.Asn36Asp maintained interactions with solvent, LPA p.Tyr2023Cys reduced intramolecular interaction with Gln1987, and KIF6 p.Trp719Arg did not affect intramolecular interactions. DDMut analysis showed that LPA p.Arg2016Cys and p.Tyr2023Cys and LPL p.Asp36Asn caused energetically favorable changes, and KIF6 p.Trp719Arg resulted in unfavorable energetic changes, affecting protein stability. CONCLUSION Deleterious variants in ABCA1, LPA, LPL and KIF6 are associated with variability in LDLc response to statins, and ABCA1 rs769705621 is associated with SRAE risk in FH patients. Molecular modeling studies suggest that LPA p.Tyr2023Cys and KIF6 p.Trp719Arg disturb protein conformational structure and stability.
Collapse
Affiliation(s)
- Carolina Dagli-Hernandez
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of Sao Paulo, Sao Paulo, Brazil
| | - Glaucio Monteiro Ferreira
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of Sao Paulo, Sao Paulo, Brazil
| | - Renata Caroline Costa de Freitas
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of Sao Paulo, Sao Paulo, Brazil
- Department of Cardiac Surgery, Boston Children's Hospital, Boston, Massachusetts, USA
| | | | - Victor Fernandes de Oliveira
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of Sao Paulo, Sao Paulo, Brazil
| | | | | | | | | | - Raul Hernandes Bortolin
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of Sao Paulo, Sao Paulo, Brazil
- Department of Cardiology, Boston Children's Hospital, Boston, Massachusetts, USA
| | - Mario Hiroyuki Hirata
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of Sao Paulo, Sao Paulo, Brazil
| | - Rosario Dominguez Crespo Hirata
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of Sao Paulo, Sao Paulo, Brazil
| |
Collapse
|
20
|
Tian X, Zhang N, Tse G, Li G, Sun Y, Liu T. Association between lipoprotein(a) and premature atherosclerotic cardiovascular disease: a systematic review and meta-analysis. EUROPEAN HEART JOURNAL OPEN 2024; 4:oeae031. [PMID: 38737415 PMCID: PMC11086656 DOI: 10.1093/ehjopen/oeae031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Revised: 03/15/2024] [Accepted: 03/26/2024] [Indexed: 05/14/2024]
Abstract
AIMS High lipoprotein(a) [Lp(a)] level has been demonstrated as an important risk factor for atherosclerotic cardiovascular diseases (ASCVD) amongst the older populations, whereas its effects in the younger population remain unclear. This study evaluated the associations between Lp(a) and the risk of premature ASCVD. METHOD AND RESULTS PubMed and Embase were searched for related studies until 12 November 2023. Fifty-one studies including 100 540 participants were included. Mean age of patients ranged from 35.3 to 62.3 years. The proportion of male participants ranged from 0% to 100%. The mean follow-up was provided in five studies ranging from 1 year to 40 years. The definition of elevated Lp(a) varied among studies, such as >30 mg/dL, >50 mg/dL, the top tertiles, the top quartiles, the top quintiles, and so on. Higher Lp(a) was significantly associated with the composite ASCVD [odds ratio (OR): 2.15, 95% confidence interval (95% CI): 1.53-3.02, P < 0.001], especially for coronary artery disease (OR: 2.44, 95% CI: 2.06-2.90, P < 0.001) and peripheral arterial disease (OR: 2.56, 95% CI: 1.56-4.21, P < 0.001). This association remained significant in familial hypercholesterolaemia (FH) (OR: 3.11, 95% CI: 1.63-5.96, P < 0.001) and type 2 diabetes mellitus (T2DM) patients (OR: 2.23; 95% CI: 1.54-3.23, P < 0.001).Significant results were observed in South Asians (OR: 3.71, 95% CI: 2.31-5.96, P < 0.001), Caucasians (OR: 3.17, 95% CI: 2.22-4.52, P < 0.001), and patients with baseline low-density lipoprotein cholesterol (LDL-c) level ≥ 2.6 mmol/L. CONCLUSION Elevated Lp(a) predicts the risk of the composite or individual ASCVD in young, regardless of study design, gender, population characteristics (community or hospitalized), different premature definitions, and various Lp(a) measurement approaches. This association was important in South Asians, Caucasians, FH patients, T2DM patients, and patients with baseline LDL-c level ≥ 2.6 mmol/L.
Collapse
Affiliation(s)
- Xu Tian
- Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular Disease, Department of Cardiology, Tianjin Institute of Cardiology, Second Hospital of Tianjin Medical University, Tianjin 300211, China
| | - Nan Zhang
- Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular Disease, Department of Cardiology, Tianjin Institute of Cardiology, Second Hospital of Tianjin Medical University, Tianjin 300211, China
| | - Gary Tse
- Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular Disease, Department of Cardiology, Tianjin Institute of Cardiology, Second Hospital of Tianjin Medical University, Tianjin 300211, China
- School of Nursing and Health Studies, Hong Kong Metropolitan University, Hong Kong, China
- Epidemiology Research Unit, Cardiovascular Analytics Group, PowerHealth Limited, Hong Kong, China
| | - Guangping Li
- Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular Disease, Department of Cardiology, Tianjin Institute of Cardiology, Second Hospital of Tianjin Medical University, Tianjin 300211, China
| | - Yihong Sun
- Cardiology Department, China-Japan Friendship Hospital, Beijing, China
| | - Tong Liu
- Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular Disease, Department of Cardiology, Tianjin Institute of Cardiology, Second Hospital of Tianjin Medical University, Tianjin 300211, China
| |
Collapse
|
21
|
Ferri N, Ruscica M, Fazio S, Corsini A. Low-Density Lipoprotein Cholesterol-Lowering Drugs: A Narrative Review. J Clin Med 2024; 13:943. [PMID: 38398257 PMCID: PMC10889346 DOI: 10.3390/jcm13040943] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2024] [Revised: 01/26/2024] [Accepted: 02/02/2024] [Indexed: 02/25/2024] Open
Abstract
The modern history of cholesterol-lowering drugs started in 1972 when Dr. Akira Endo identified an active compound (compactin) that inhibited cholesterol biosynthesis from the culture broth of blue-green mold (Penicillium citrinum Pen-51). Since 1987, statins have represented the milestone for the treatment of atherosclerotic cardiovascular disease. A new therapy for the treatment of hypercholesterolemia since the discovery of statins is ezetimibe, the first and only agent inhibiting intestinal cholesterol absorption. Ezetimibe was approved by the FDA in October 2002. A year later, the association between gain-of-function PCSK9 genetic mutations and hypercholesterolemia was reported, and this discovery opened a new era in lipid-lowering therapies. Monoclonal antibodies and small-interfering RNA approaches to reduce PCSK9 were developed and approved for clinical use in 2015 and 2022, respectively. Finally, the newly approved bempedoic acid, an oral adenosine triphosphate citrate lyase inhibitor that lowers LDL-C, is able to reduce major adverse cardiovascular events in both primary and secondary prevention. In the present narrative review, we summarize the pharmacological properties and the clinical efficacy of all these agents currently used for a tailored therapy of hypercholesterolemia in patients with atherosclerotic cardiovascular disease.
Collapse
Affiliation(s)
- Nicola Ferri
- Department of Medicine (DIMED), University of Padova, 35122 Padova, Italy
- Veneto Institute of Molecular Medicine (VIMM), 35129 Padua, Italy
| | - Massimiliano Ruscica
- Department of Pharmacological and Biomolecular Sciences “Rodolfo Paoletti”, University of Milan, 20133 Milan, Italy;
- Department of Cardio-Thoracic-Vascular Diseases, Foundation IRCCS Cà Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy
| | - Sergio Fazio
- Regeneron Pharmaceuticals Inc., Tarrytown, NY 10591, USA;
| | - Alberto Corsini
- Department of Pharmacological and Biomolecular Sciences “Rodolfo Paoletti”, University of Milan, 20133 Milan, Italy;
| |
Collapse
|
22
|
Kamili K, Zheng T, Luo C, Wang X, Tian G. Predictive value of lipoprotein(a) for left atrial thrombus or spontaneous echo contrast in non-valvular atrial fibrillation patients with low CHA 2DS 2-VASc scores: a cross-sectional study. Lipids Health Dis 2024; 23:22. [PMID: 38254171 PMCID: PMC10801929 DOI: 10.1186/s12944-023-01990-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Accepted: 12/24/2023] [Indexed: 01/24/2024] Open
Abstract
OBJECTIVE Current guidelines are debated when it comes to starting anticoagulant therapy in patients with non-valvular atrial fibrillation (NVAF) and low CHA2DS2-VASc scores (1-2 in women and 0-1 in men). However, these individuals still have a high likelihood of developing left atrial thrombus/spontaneous echo contrast (LAT/SEC) and experiencing subsequent thromboembolism. Recent research has demonstrated that lipoprotein(a) [Lp(a)] may increase the risk of thrombosis, but the relationship between Lp(a) and LAT/SEC in NVAF patients is not clearly established. Therefore, this study sought to evaluate the predictive ability of Lp(a) for LAT/SEC among NVAF patients with low CHA2DS2-VASc scores. METHODS NVAF patients with available transesophageal echocardiography (TEE) data were evaluated. Based on the TEE results, the subjects were classified into non-LAT/SEC and LAT/SEC groups. The risk factors for LAT/SEC were examined using binary logistic regression analyses and were validated by using 1:1 propensity score matching (PSM). Subsequently, novel predictive models for LAT/SEC were developed by integrating the CHA2DS2-VASc score with the identified factors, and the accuracy of these models was tested using receiver operating characteristic (ROC) analysis. RESULTS In total, 481 NVAF patients were enrolled. The LAT/SEC group displayed higher Lp(a) concentrations. It was found that enlarged left atrial diameter (LAD), high concentrations of Lp(a), and a history of coronary heart disease (CHD) were independent predictors of LAT/SEC. Lp(a) and LAD still had predictive values for LAT/SEC after adjusting for PSM. In both the highest quartile groups of Lp(a) (>266 mg/L) and LAD (>39.5 mm), the occurrence of LAT/SEC was higher than that in the corresponding lowest quartile. By incorporating Lp(a) and the LAD, the predictive value of the CHA2DS2-VASc score for LAT/SEC was significantly improved. CONCLUSION Elevated Lp(a) and enlarged LAD were independent risk factors for LAT/SEC among NVAF patients with low CHA2DS2-VASc scores. The prediction accuracy of the CHA2DS2-VASc score for LAT/SEC was significantly improved by the addition of Lp(a) and LAD. When evaluating the stroke risk in patients with NVAF, Lp(a) and LAD should be taken into account together with the CHA2DS2-VASc score. TRIAL REGISTRATION Retrospectively registered.
Collapse
Affiliation(s)
- Kamila Kamili
- Department of Cardiovascular Medicine, First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta West Rd, Xi'an, Shaanxi, 710061, People's Republic of China
| | - Tingting Zheng
- Department of Cardiovascular Medicine, First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta West Rd, Xi'an, Shaanxi, 710061, People's Republic of China
| | - Chaodi Luo
- Department of Cardiovascular Medicine, First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta West Rd, Xi'an, Shaanxi, 710061, People's Republic of China
| | - Xuan Wang
- Department of Cardiovascular Medicine, First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta West Rd, Xi'an, Shaanxi, 710061, People's Republic of China
| | - Gang Tian
- Department of Cardiovascular Medicine, First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta West Rd, Xi'an, Shaanxi, 710061, People's Republic of China.
| |
Collapse
|
23
|
Heydari M, Rezayi M, Ruscica M, Jpamialahamdi T, Johnston TP, Sahebkar A. The ins and outs of lipoprotein(a) assay methods. Arch Med Sci Atheroscler Dis 2023; 8:e128-e139. [PMID: 38283929 PMCID: PMC10811544 DOI: 10.5114/amsad/176653] [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: 11/11/2023] [Accepted: 12/08/2023] [Indexed: 01/30/2024] Open
Abstract
Pathophysiological, epidemiological and genetic studies convincingly showed lipoprotein(a) (Lp(a)) to be a causal mediator of atherosclerotic cardiovascular disease (ASCVD). This happens through a myriad of mechanisms including activation of innate immune cells, endothelial cells as well as platelets. Although these certainties whether or not Lp(a) is ready for prime-time clinical use remain debated. Thus, remit of the present review is to provide an overview of different methods that have been employed for the measurement of Lp(a). The methods include dynamic light scattering, multi-angle light scattering analysis, near-field imaging, sedimentation, gel filtration, and electron microscopy. The development of multiple Lp(a) detection methods is vital for improved prediction of ASCVD risk.
Collapse
Affiliation(s)
- Maryam Heydari
- Medical Toxicology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Majid Rezayi
- Medical Toxicology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Metabolic Syndrome Research Center, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Medical Biotechnology and Nanotechnology, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Massimiliano Ruscica
- Department of Pharmacological and Biomolecular Science, Università degli Studi di Milano, Milan, Italy
| | - Tannaz Jpamialahamdi
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Thomas P. Johnston
- Division of Pharmacology and Pharmaceutical Science, School of Pharmacy, University of Missouri-Kansas City, Kansas City, MI, USA
| | - Amirhossein Sahebkar
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
- School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
24
|
Carugo S, Sirtori CR, Gelpi G, Corsini A, Tokgozoglu L, Ruscica M. Updates in Small Interfering RNA for the Treatment of Dyslipidemias. Curr Atheroscler Rep 2023; 25:805-817. [PMID: 37792132 PMCID: PMC10618314 DOI: 10.1007/s11883-023-01156-5] [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] [Accepted: 09/17/2023] [Indexed: 10/05/2023]
Abstract
PURPOSE OF REVIEW Atherosclerotic cardiovascular disease (ASCVD) is still the leading cause of death worldwide. Despite excellent pharmacological approaches, clinical registries consistently show that many people with dyslipidemia do not achieve optimal management, and many of them are treated with low-intensity lipid-lowering therapies. Beyond the well-known association between low-density lipoprotein cholesterol (LDL-C) and cardiovascular prevention, the atherogenicity of lipoprotein(a) and the impact of triglyceride (TG)-rich lipoproteins cannot be overlooked. Within this landscape, the use of RNA-based therapies can help the treatment of difficult to target lipid disorders. RECENT FINDINGS The safety and efficacy of LDL-C lowering with the siRNA inclisiran has been documented in the open-label ORION-3 trial, with a follow-up of 4 years. While the outcome trial is pending, a pooled analysis of ORION-9, ORION-10, and ORION-11 has shown the potential of inclisiran to reduce composite major adverse cardiovascular events. Concerning lipoprotein(a), data of OCEAN(a)-DOSE trial with olpasiran show a dose-dependent drop in lipoprotein(a) levels with an optimal pharmacodynamic profile when administered every 12 weeks. Concerning TG lowering, although ARO-APOC3 and ARO-ANG3 are effective to lower apolipoprotein(apo)C-III and angiopoietin-like 3 (ANGPTL3) levels, these drugs are still in their infancy. In the era moving toward a personalized risk management, the use of siRNA represents a blossoming armamentarium to tackle dyslipidaemias for ASCVD risk reduction.
Collapse
Affiliation(s)
- S Carugo
- Department of Clinical Sciences and Community Health, Dyspnea Lab, Università degli Studi di Milano, Milan, Italy
- Department of Cardio-Thoracic-Vascular Diseases - Foundation IRCCS Cà Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - C R Sirtori
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Milan, Italy
| | - G Gelpi
- Department of Cardio-Thoracic-Vascular Diseases - Foundation IRCCS Cà Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - A Corsini
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Milan, Italy
| | - L Tokgozoglu
- Department of Cardiology, Hacettepe University Faculty of Medicine, Ankara, Turkey
| | - M Ruscica
- Department of Cardio-Thoracic-Vascular Diseases - Foundation IRCCS Cà Granda Ospedale Maggiore Policlinico, Milan, Italy.
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Milan, Italy.
| |
Collapse
|
25
|
Wang H, Fan JL. Association between lipoprotein(a) and insulin resistance in Chinese adults: results from the China health and nutrition survey. Front Endocrinol (Lausanne) 2023; 14:1234140. [PMID: 37876542 PMCID: PMC10591330 DOI: 10.3389/fendo.2023.1234140] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/03/2023] [Accepted: 09/26/2023] [Indexed: 10/26/2023] Open
Abstract
Background Lipoprotein(a) [Lp(a)] is a well-established risk factor for cardiovascular diseases. However, the relationship between Lp(a) and insulin resistance (IR) remains controversial. The aim of the current study was to investigate the association between Lp(a) concentrations and IR in Chinese adults. Methods Cross-sectional study of 1908 cases and 5725 controls was performed for identifying the association of Lp(a) with IR. IR was assessed using the triglyceride glucose (TyG) index, and patients with a TyG index greater than the third quartile were defined as having IR. Results The distribution of Lp(a) in Chinese adults was skewed, with a median of 7.90mg/dL. Lp(a) concentrations were significantly and progressively lower with increasing TyG index values in Chinese adult males, but not in females. Multiple regression analysis adjusted for a wide range of risk factors showed that Lp(a) concentrations were inversely and independently associated with IR in Chinese adult males, but not in females. The suggested Lp (a) cutoff for discriminating IR from non-IR was 4.7 mg/dL in Chinese adult males. Lp(a) interacts with gender in IR on both additive and multiplicative scale in Chinese adults. Conclusion Lp(a) concentrations inversely associated with IR in Chinses adult males, but the association in women needs further study. In Chinese adults, Lp(a) interacts with gender in IR.
Collapse
Affiliation(s)
| | - Jia-Li Fan
- Division of Cardiology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| |
Collapse
|
26
|
Koschinsky ML, Stroes ESG, Kronenberg F. Daring to dream: Targeting lipoprotein(a) as a causal and risk-enhancing factor. Pharmacol Res 2023; 194:106843. [PMID: 37406784 DOI: 10.1016/j.phrs.2023.106843] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Revised: 06/15/2023] [Accepted: 06/27/2023] [Indexed: 07/07/2023]
Abstract
Lipoprotein(a) [Lp(a)], a distinct lipoprotein class, has become a major focus for cardiovascular research. This review is written in light of the recent guideline and consensus statements on Lp(a) and focuses on 1) the causal association between Lp(a) and cardiovascular outcomes, 2) the potential mechanisms by which elevated Lp(a) contributes to cardiovascular diseases, 3) the metabolic insights on the production and clearance of Lp(a) and 4) the current and future therapeutic approaches to lower Lp(a) concentrations. The concentrations of Lp(a) are under strict genetic control. There exists a continuous relationship between the Lp(a) concentrations and risk for various endpoints of atherosclerotic cardiovascular disease (ASCVD). One in five people in the Caucasian population is considered to have increased Lp(a) concentrations; the prevalence of elevated Lp(a) is even higher in black populations. This makes Lp(a) a cardiovascular risk factor of major public health relevance. Besides the association between Lp(a) and myocardial infarction, the relationship with aortic valve stenosis has become a major focus of research during the last decade. Genetic studies provided strong support for a causal association between Lp(a) and cardiovascular outcomes: carriers of genetic variants associated with lifelong increased Lp(a) concentration are significantly more frequent in patients with ASCVD. This has triggered the development of drugs that can specifically lower Lp(a) concentrations: mRNA-targeting therapies such as anti-sense oligonucleotide (ASO) therapies and short interfering RNA (siRNA) therapies have opened new avenues to lower Lp(a) concentrations more than 95%. Ongoing Phase II and III clinical trials of these compounds are discussed in this review.
Collapse
Affiliation(s)
- Marlys L Koschinsky
- Robarts Research Institute, Schulich School of Medicine & Dentistry, The University of Western Ontario, London, Ontario, Canada; Department of Physiology & Pharmacology, Schulich School of Medicine & Dentistry, The University of Western Ontario, London, Ontario, Canada
| | - Erik S G Stroes
- Department of Vascular Medicine, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
| | - Florian Kronenberg
- Institute of Genetic Epidemiology, Medical University of Innsbruck, Innsbruck, Austria.
| |
Collapse
|
27
|
Serra-Planas E. Referral rate, profile and degree of control of patients with familial hypercholesterolemia: data from a single lipid unit from a Mediterranean area. Lipids Health Dis 2023; 22:62. [PMID: 37170237 PMCID: PMC10176814 DOI: 10.1186/s12944-023-01815-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2023] [Accepted: 04/10/2023] [Indexed: 05/13/2023] Open
Abstract
BACKGROUND The challenging rigorous management of hypercholesterolemia promotes referral to specialized units. This study explored the need, based on referral rate and cardiovascular (CV) risk factor control in patients evaluated for familial hypercholesterolemia (FH), for a lipid unit (LU). METHODS Over a four-year period, 340 referrals to our unit were analyzed to establish the lipid disorder referral rate. Moreover, 118 patients referred for potential FH during the period 2010-2018 (52.4 ± 13.9 years, 47.5% male, Caucasian, 26.3% obese, 33.1% smokers and 51.7% with some glycaemic alteration) were investigated. The Dutch Lipid Clinic Network (DLCN) score, type and dose of lipid-lowering drugs, lipid profile including lipoprotein (a) (Lp(a)) and the presence of plaques with carotid ultrasound (CU) were recorded. RESULTS Lipids represented 6.2% of referrals (38 patient-years) requiring a 2-3 h weekly monographic outpatient consultation. The potential FH sample displayed a DLCN score ≥ 6 in 78% and modifiable CV risk factors in 51%. Only 22% achieved tight disease control despite intensive treatment. The statin-ezetimibe combination treatment group achieved better goals (73.0% vs. 45.5%, P = 0.003), and the rosuvastatin group had a higher proportion of prediabetes (60.9% vs. 39.1%, P = 0.037). Neither CU plaque presence nor Lp(a) > 50 mg/dL was linked with established CV disease patients, but higher Lp(a) concentrations were detected between them (102.5 (26.3-145.8) vs. 25.0 (13.0-52.0) mg/dL, P = 0.012). CONCLUSIONS The referral rate, degree of control, and proportion of modifiable CV risk factors in FH patients demonstrate the need for LU in our area as well as optimize control and treatment.
Collapse
Affiliation(s)
- Enric Serra-Planas
- Department of Endocrinology, Internal Medicine Service, Hospital Universitari d'Igualada, Consorci Sanitari de l'Anoia, Avinguda de Catalunya, 11, Igualada, Barcelona, 08700, Spain.
- Unit of lipids and cardiovascular risk, University Hospital of Igualada, Barcelona, Spain.
| |
Collapse
|
28
|
Piccioni A, Niccolai E, Rozzi G, Spaziani G, Zanza C, Candelli M, Covino M, Gasbarrini A, Franceschi F, Amedei A. Familial Hypercholesterolemia and Acute Coronary Syndromes: The Microbiota-Immunity Axis in the New Diagnostic and Prognostic Frontiers. Pathogens 2023; 12:627. [PMID: 37111513 PMCID: PMC10142551 DOI: 10.3390/pathogens12040627] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Revised: 04/18/2023] [Accepted: 04/20/2023] [Indexed: 04/29/2023] Open
Abstract
Familial hypercholesterolemia is a common genetic disorder with a propensity towards early onset of atherosclerotic cardiovascular disease (CVD). The main goal of therapy is to reduce the LDL cholesterol and the current treatment generally consists of statin, ezetimibe and PCSK9 inhibitors. Unfortunately, lowering LDL cholesterol may be difficult for many reasons such as the variation of response to statin therapy among the population or the high cost of some therapies (i.e., PCSK9 inhibitors). In addition to conventional therapy, additional strategies may be used. The gut microbiota has been recently considered to play a part in chronic systemic inflammation and hence in CVD. Several studies, though they are still preliminary, consider dysbiosis a risk factor for various CVDs through several mechanisms. In this review, we provide an update of the current literature about the intricate relation between the gut microbiota and the familial hypercholesterolemia.
Collapse
Affiliation(s)
- Andrea Piccioni
- Emergency Medicine Department, Fondazione Policlinico Universitario Agostino Gemelli-IRCCS, Università Cattolica del Sacro Cuore di Roma, 00168 Roma, Italy
| | - Elena Niccolai
- Department of Experimental and Clinical Medicine, University of Florence, 50134 Florence, Italy
| | - Gloria Rozzi
- Emergency Medicine Department, Fondazione Policlinico Universitario Agostino Gemelli-IRCCS, Università Cattolica del Sacro Cuore di Roma, 00168 Roma, Italy
| | - Giacomo Spaziani
- Emergency Medicine Department, Fondazione Policlinico Universitario Agostino Gemelli-IRCCS, Università Cattolica del Sacro Cuore di Roma, 00168 Roma, Italy
| | - Christian Zanza
- Foundation “Ospedale Alba-Bra Onlus”, Department of Emergency Medicine, Anesthesia and Critical Care Medicine, Michele and Pietro Ferrero Hospital, 12060 Verduno, Italy
| | - Marcello Candelli
- Emergency Medicine Department, Fondazione Policlinico Universitario Agostino Gemelli-IRCCS, Università Cattolica del Sacro Cuore di Roma, 00168 Roma, Italy
| | - Marcello Covino
- Emergency Medicine Department, Fondazione Policlinico Universitario Agostino Gemelli-IRCCS, Università Cattolica del Sacro Cuore di Roma, 00168 Roma, Italy
| | - Antonio Gasbarrini
- Medical and Surgical Science Department, Fondazione Policlinico Universitario Agostino Gemelli-IRCCS, Università Cattolica del Sacro Cuore di Roma, 00168 Roma, Italy
| | - Francesco Franceschi
- Emergency Medicine Department, Fondazione Policlinico Universitario Agostino Gemelli-IRCCS, Università Cattolica del Sacro Cuore di Roma, 00168 Roma, Italy
| | - Amedeo Amedei
- Department of Experimental and Clinical Medicine, University of Florence, 50134 Florence, Italy
| |
Collapse
|
29
|
Hussain Z, Iqbal J, Liu H, Zhou HD. Exploring the role of lipoprotein(a) in cardiovascular diseases and diabetes in Chinese population. Int J Biol Macromol 2023; 233:123586. [PMID: 36758756 DOI: 10.1016/j.ijbiomac.2023.123586] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Revised: 01/31/2023] [Accepted: 02/04/2023] [Indexed: 02/10/2023]
Abstract
A high level of lipoprotein (a) in the plasma has been associated with a variety of cardiovascular diseases and is considered to be an independent predictor of some other diseases. Based on recent studies, the concentration levels of Lp(a) in the Chinese population exhibit a distinctive variation from other populations. In the Chinese population, a high level of Lp(a) indicates a higher incidence of revascularization, platelet aggregation, and thrombogenicity following PCI. Increased risk of atherosclerotic cardiovascular disease (ASCVD) in Chinese population has been linked to higher levels of Lp(a), according to studies. More specifically, it has been found that in Chinese populations, higher levels of Lp(a) were linked to an increased risk of coronary heart disease, severe aortic valve stenosis, deep vein thrombosis in patients with spinal cord injuries, central vein thrombosis in patients receiving hemodialysis, and stroke. Furthermore, new and consistent data retrieved from several clinical trials also suggest that Lp (a) might also play an essential role in some other conditions, including metabolic syndrome, type 2 diabetes and cancers. This review explores the clinical and epidemiological relationships among Lp(a), cardiovascular diseases and diabetes in the Chinese population as well as potential Lp(a) underlying mechanisms in these diseases. However, further research is needed to better understand the role of Lp(a) in cardiovascular diseases and especially diabetes in the Chinese population.
Collapse
Affiliation(s)
- Zubair Hussain
- State Key Laboratory of Esophageal Cancer Prevention and Treatment, Zhengzhou University, 450000, Zhengzhou, China; Department of Pathophysiology, Academy of Medical Science, College of Medicine, Zhengzhou University, 450000, Zhengzhou, China; China-US (Henan) Hormel Cancer Institute, 450000, Zhengzhou, China
| | - Junaid Iqbal
- National Clinical Research Center for Metabolic Diseases, Hunan Provincial Key Laboratory for Metabolic Bone Diseases, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha 410011, China
| | - Hongcai Liu
- Institute of Systems Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100005, China; Suzhou Institute of Systems Medicine, Suzhou 215123, China
| | - Hou-De Zhou
- National Clinical Research Center for Metabolic Diseases, Hunan Provincial Key Laboratory for Metabolic Bone Diseases, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha 410011, China.
| |
Collapse
|
30
|
Lin CY, Hsu SHJ, Chen CW, Wang C, Sung FC, Su TC. Association of Urinary Lead and Cadmium Levels, and Serum Lipids with Subclinical Arteriosclerosis: Evidence from Taiwan. Nutrients 2023; 15:nu15030571. [PMID: 36771277 PMCID: PMC9919350 DOI: 10.3390/nu15030571] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2022] [Revised: 01/14/2023] [Accepted: 01/16/2023] [Indexed: 01/24/2023] Open
Abstract
BACKGROUND Exposure to lead and cadmium has been linked to changes in lipid metabolism and the development of arteriosclerosis, but the role of lipoprotein profiles in this relationship is not well understood, including the potential role of novel lipid biomarkers. METHODS In this study, we enrolled 736 young Taiwanese subjects aged 12 to 30 years to assess the correlation between urine levels of lead and cadmium, lipoprotein profiles, and carotid intima-media thickness (CIMT). RESULTS Higher levels of lead and cadmium were significantly associated with higher levels of low-density lipoprotein cholesterol (LDL-C), small dense LDL-C (sdLDL-C), LDL-triglyceride (LDL-TG), and CIMT. Participants with higher levels of lead and cadmium had the highest mean values of CIMT, LDL-C, sdLDL-C, and LDL-TG. In a structural equation model, lead had a direct and indirect association with CIMT through LDL-C and sdLDL-C, whereas cadmium had a direct association with CIMT and an indirect association through LDL-C. CONCLUSION Our results suggest higher levels of lead and cadmium are associated with abnormal lipid profiles and increased CIMT. These heavy metals could have additive effects on lipids and CIMT, and the relationship between them may be mediated by lipoprotein levels. Further research is needed to determine the causal relationship.
Collapse
Affiliation(s)
- Chien-Yu Lin
- Department of Internal Medicine, En Chu Kong Hospital, New Taipei City 237, Taiwan
- School of Medicine, Fu Jen Catholic University, New Taipei City 242, Taiwan
- Department of Environmental Engineering and Health, Yuanpei University of Medical Technology, Hsinchu 300, Taiwan
| | - Sandy Huey-Jen Hsu
- Department of Laboratory Medicine, College of Medicine, National Taiwan University Hospital, National Taiwan University, Taipei 100, Taiwan
| | - Ching-Way Chen
- Department of Cardiology, National Taiwan University Hospital, Yunlin Branch, Yunlin 640, Taiwan
| | - Chikang Wang
- Department of Environmental Engineering and Health, Yuanpei University of Medical Technology, Hsinchu 300, Taiwan
| | - Fung-Chang Sung
- Department of Health Services Administration, College of Public Health, China Medical University, Taichung 404, Taiwan
- Department of Food Nutrition and Health Biotechnology, Asia University, Taichung 413, Taiwan
| | - Ta-Chen Su
- Department of Environmental and Occupational Medicine, National Taiwan University Hospital, Taipei 100, Taiwan
- Department of Internal Medicine and Cardiovascular Center, National Taiwan University Hospital, Taipei 100, Taiwan
- Institute of Environmental and Occupational Health Sciences, College of Public Health, National Taiwan University, Taipei 100, Taiwan
- The Experimental Forest, National Taiwan University, Nantou 558, Taiwan
- Correspondence: ; Tel.: +886-2-23123456 (ext. 67183); Fax: +886-2-23712361
| |
Collapse
|
31
|
Lampsas S, Xenou M, Oikonomou E, Pantelidis P, Lysandrou A, Sarantos S, Goliopoulou A, Kalogeras K, Tsigkou V, Kalpis A, Paschou SA, Theofilis P, Vavuranakis M, Tousoulis D, Siasos G. Lipoprotein(a) in Atherosclerotic Diseases: From Pathophysiology to Diagnosis and Treatment. Molecules 2023; 28:969. [PMID: 36770634 PMCID: PMC9918959 DOI: 10.3390/molecules28030969] [Citation(s) in RCA: 53] [Impact Index Per Article: 26.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Revised: 01/12/2023] [Accepted: 01/17/2023] [Indexed: 01/21/2023] Open
Abstract
Lipoprotein(a) (Lp(a)) is a low-density lipoprotein (LDL) cholesterol-like particle bound to apolipoprotein(a). Increased Lp(a) levels are an independent, heritable causal risk factor for atherosclerotic cardiovascular disease (ASCVD) as they are largely determined by variations in the Lp(a) gene (LPA) locus encoding apo(a). Lp(a) is the preferential lipoprotein carrier for oxidized phospholipids (OxPL), and its role adversely affects vascular inflammation, atherosclerotic lesions, endothelial function and thrombogenicity, which pathophysiologically leads to cardiovascular (CV) events. Despite this crucial role of Lp(a), its measurement lacks a globally unified method, and, between different laboratories, results need standardization. Standard antilipidemic therapies, such as statins, fibrates and ezetimibe, have a mediocre effect on Lp(a) levels, although it is not yet clear whether such treatments can affect CV events and prognosis. This narrative review aims to summarize knowledge regarding the mechanisms mediating the effect of Lp(a) on inflammation, atherosclerosis and thrombosis and discuss current diagnostic and therapeutic potentials.
Collapse
Affiliation(s)
- Stamatios Lampsas
- 3rd Department of Cardiology, National and Kapodistrian University of Athens, Medical School, Sotiria Chest Disease Hospital, 11527 Athens, Greece
| | - Maria Xenou
- 3rd Department of Cardiology, National and Kapodistrian University of Athens, Medical School, Sotiria Chest Disease Hospital, 11527 Athens, Greece
| | - Evangelos Oikonomou
- 3rd Department of Cardiology, National and Kapodistrian University of Athens, Medical School, Sotiria Chest Disease Hospital, 11527 Athens, Greece
| | - Panteleimon Pantelidis
- 3rd Department of Cardiology, National and Kapodistrian University of Athens, Medical School, Sotiria Chest Disease Hospital, 11527 Athens, Greece
| | - Antonios Lysandrou
- 3rd Department of Cardiology, National and Kapodistrian University of Athens, Medical School, Sotiria Chest Disease Hospital, 11527 Athens, Greece
| | - Savvas Sarantos
- 3rd Department of Cardiology, National and Kapodistrian University of Athens, Medical School, Sotiria Chest Disease Hospital, 11527 Athens, Greece
| | - Athina Goliopoulou
- 3rd Department of Cardiology, National and Kapodistrian University of Athens, Medical School, Sotiria Chest Disease Hospital, 11527 Athens, Greece
| | - Konstantinos Kalogeras
- 3rd Department of Cardiology, National and Kapodistrian University of Athens, Medical School, Sotiria Chest Disease Hospital, 11527 Athens, Greece
- 1st Department of Cardiology, National and Kapodistrian University of Athens, Medical School, Hippokration General Hospital, 11527 Athens, Greece
| | - Vasiliki Tsigkou
- 3rd Department of Cardiology, National and Kapodistrian University of Athens, Medical School, Sotiria Chest Disease Hospital, 11527 Athens, Greece
| | - Athanasios Kalpis
- 3rd Department of Cardiology, National and Kapodistrian University of Athens, Medical School, Sotiria Chest Disease Hospital, 11527 Athens, Greece
| | - Stavroula A. Paschou
- 1st Department of Cardiology, National and Kapodistrian University of Athens, Medical School, Hippokration General Hospital, 11527 Athens, Greece
| | - Panagiotis Theofilis
- 3rd Department of Cardiology, National and Kapodistrian University of Athens, Medical School, Sotiria Chest Disease Hospital, 11527 Athens, Greece
| | - Manolis Vavuranakis
- 3rd Department of Cardiology, National and Kapodistrian University of Athens, Medical School, Sotiria Chest Disease Hospital, 11527 Athens, Greece
| | - Dimitris Tousoulis
- 1st Department of Cardiology, National and Kapodistrian University of Athens, Medical School, Hippokration General Hospital, 11527 Athens, Greece
| | - Gerasimos Siasos
- 3rd Department of Cardiology, National and Kapodistrian University of Athens, Medical School, Sotiria Chest Disease Hospital, 11527 Athens, Greece
- Cardiovascular Division, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
32
|
Raitakari O, Kartiosuo N, Pahkala K, Hutri-Kähönen N, Bazzano LA, Chen W, Urbina EM, Jacobs DR, Sinaiko A, Steinberger J, Burns T, Daniels SR, Venn A, Woo JG, Dwyer T, Juonala M, Viikari J. Lipoprotein(a) in Youth and Prediction of Major Cardiovascular Outcomes in Adulthood. Circulation 2023; 147:23-31. [PMID: 36440577 PMCID: PMC9797445 DOI: 10.1161/circulationaha.122.060667] [Citation(s) in RCA: 41] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Accepted: 10/17/2022] [Indexed: 11/29/2022]
Abstract
BACKGROUND Elevated lipoprotein(a) [Lp(a)] is a common risk factor for cardiovascular disease outcomes with unknown mechanisms. We examined its potential role in identifying youths who are at increased risk of developing adult atherosclerotic cardiovascular disease (ASCVD). METHODS Lp(a) levels measured in youth 9 to 24 years of age were linked to adult ASCVD and carotid intima-media thickness in the YFS (Cardiovascular Risk in Young Finns Study), in which 95 of the original 3596 participants (2.7%) recruited as children have been diagnosed with ASCVD at a median of 47 years of age. Results observed in YFS were replicated with the use of data for White participants from the BHS (Bogalusa Heart Study). In BHS, 587 White individuals had data on youth Lp(a) (measured at 8-17 years of age) and information on adult events, including 15 cases and 572 noncases. Analyses were performed with the use of Cox proportional hazard regression. RESULTS In YFS, those who had been exposed to high Lp(a) level in youth [defined as Lp(a) ≥30 mg/dL] had ≈2 times greater risk of developing adult ASCVD compared with nonexposed individuals (hazard ratio, 2.0 [95% CI, 1.4-2.6]). Youth risk factors, including Lp(a), low-density lipoprotein cholesterol, body mass index, and smoking, were all independently associated with higher risk. In BHS, in an age- and sex-adjusted model, White individuals who had been exposed to high Lp(a) had 2.5 times greater risk (95% CI, 0.9-6.8) of developing adult ASCVD compared with nonexposed individuals. When also adjusted for low-density lipoprotein cholesterol and body mass index, the risk associated with high Lp(a) remained unchanged (hazard ratio, 2.4 [95% CI, 0.8-7.3]). In a multivariable model for pooled data, individuals exposed to high Lp(a) had 2.0 times greater risk (95% CI, 1.0-3.7) of developing adult ASCVD compared with nonexposed individuals. No association was detected between youth Lp(a) and adult carotid artery thickness in either cohort or pooled data. CONCLUSIONS Elevated Lp(a) level identified in youth is a risk factor for adult atherosclerotic cardiovascular outcomes but not for increased carotid intima-media thickness.
Collapse
Affiliation(s)
- Olli Raitakari
- Centre for Population Health Research, University of Turku and Turku University Hospital, Finland (O.R., N.K., K.P.)
- Research Centre of Applied and Preventive Cardiovascular Medicine (O.R., N.K., K.P.), University of Turku, Finland
- Department of Clinical Physiology and Nuclear Medicine (O.R.), Turku University Hospital, Finland
| | - Noora Kartiosuo
- Centre for Population Health Research, University of Turku and Turku University Hospital, Finland (O.R., N.K., K.P.)
- Research Centre of Applied and Preventive Cardiovascular Medicine (O.R., N.K., K.P.), University of Turku, Finland
| | - Katja Pahkala
- Centre for Population Health Research, University of Turku and Turku University Hospital, Finland (O.R., N.K., K.P.)
- Research Centre of Applied and Preventive Cardiovascular Medicine (O.R., N.K., K.P.), University of Turku, Finland
- Paavo Nurmi Centre and Unit for Health and Physical Activity (K.P.), University of Turku, Finland
| | - Nina Hutri-Kähönen
- Tampere Centre for Skills Training and Simulation, Tampere University, Finland (N.H.-K.)
| | - Lydia A Bazzano
- Department of Epidemiology, School of Public Health and Tropical Medicine, Tulane University, New Orleans, LA (L.A.B., W.C.)
| | - Wei Chen
- Department of Epidemiology, School of Public Health and Tropical Medicine, Tulane University, New Orleans, LA (L.A.B., W.C.)
| | - Elaine M Urbina
- Department of Pediatrics, University of Cincinnati College of Medicine, OH (E.M.U., J.G.W.)
- The Heart Institute (E.M.U.), Cincinnati Children's Hospital Medical Center, OH
| | - David R Jacobs
- Division of Epidemiology and Community Health, School of Public Health, University of Minnesota, Minneapolis (D.R.J.)
| | - Alan Sinaiko
- Department of Pediatrics, University of Minnesota Medical School, Minneapolis (A.S., J.S.)
| | - Julia Steinberger
- Department of Pediatrics, University of Minnesota Medical School, Minneapolis (A.S., J.S.)
| | - Trudy Burns
- Department of Medicine (M.J., J.V.), University of Turku, Finland
| | - Stephen R Daniels
- Department of Pediatrics, University of Colorado School of Medicine, Aurora (S.R.D.)
- Children's Hospital Colorado, Anschutz Medical Campus, Aurora (S.R.D.)
| | - Alison Venn
- Menzies Institute for Medical Research, University of Tasmania, Hobart, Australia (A.V., T.D.)
| | - Jessica G Woo
- Department of Pediatrics, University of Cincinnati College of Medicine, OH (E.M.U., J.G.W.)
- Division of Biostatistics and Epidemiology (J.G.W.), Cincinnati Children's Hospital Medical Center, OH
| | - Terry Dwyer
- Menzies Institute for Medical Research, University of Tasmania, Hobart, Australia (A.V., T.D.)
- Heart Research Group, Murdoch Children's Research Institute, Melbourne, Australia (T.D.)
- Nuffield Department of Women's & Reproductive Health, University of Oxford, United Kingdom (T.D.)
| | - Markus Juonala
- Department of Medicine (M.J., J.V.), University of Turku, Finland
- Division of Medicine (M.J., J.V.), Turku University Hospital, Finland
| | - Jorma Viikari
- Department of Medicine (M.J., J.V.), University of Turku, Finland
| |
Collapse
|
33
|
Wang A, Zhang S, Li Y, Zhu F, Xie B. Study on the relationship between lipoprotein (a) and diabetic kidney disease. J Diabetes Complications 2023; 37:108378. [PMID: 36549039 DOI: 10.1016/j.jdiacomp.2022.108378] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Revised: 11/29/2022] [Accepted: 12/01/2022] [Indexed: 12/23/2022]
Abstract
OBJECTIVE Little is currently known about the role of lipid metabolism in diabetic kidney disease (DKD), warranting further study. The present study sought to investigate the correlation between lipid metabolism and renal function as well as renal pathological grade/score in DKD patients. METHODS A total of 224 patients diagnosed with DKD by pathological examination were retrospectively analyzed, of which 74 patients were further evaluated by DKD pathological grade/score. ANOVA was used to investigate serum lipoprotein (a) [Lp (a)] levels in DKD patients with different chronic kidney disease (CKD) stages. Spearman correlation analysis was used to evaluate the relationship between Lp (a) and renal function-related indicators. The DKD pathological grade/score was also evaluated with this method. The receiver operating characteristic (ROC) curve was used to analyze the value of Lp (a) in assessing renal function and pathological changes. RESULTS There were significant differences in Lp (a) levels among different CKD stages (H = 17.063, p = 0.002) and glomerular grades (H = 12.965, p = 0.005). Lp (a) levels correlated with serum creatinine (p = 0.000), blood urea nitrogen (p = 0.000), estimated glomerular filtration rate (p = 0.000), 24-h proteinuria (24hUPro, p = 0.000), urine microalbumin (p = 0.000), urine albumin creatinine ratio (p = 0.000), glomerular basement membrane thickness (p = 0.003), and glomerular grade (p = 0.039). ROC curve demonstrated good performance of Lp (a) as an indicator to assess CKD stage 4-5 (AUC = 0.684, p = 0.000), 24hUPro > 3.5 g (AUC = 0.720, p = 0.000), and glomerular grade III-IV (AUC = 0.695, p = 0.012). CONCLUSIONS Elevated levels of Lp (a) are associated with decreased GFR, increased proteinuria, and renal pathological progression, suggesting they could be used to monitor changes in DKD patients.
Collapse
Affiliation(s)
- Anni Wang
- Hangzhou Hospital of Traditional Chinese Medicine, Hangzhou TCM Hospital Affiliated to Zhejiang Chinese Medical University, Tiyuchang Rd 453, Hangzhou 310007, People's Republic of China.
| | - Shaojie Zhang
- Hangzhou Hospital of Traditional Chinese Medicine, Hangzhou TCM Hospital Affiliated to Zhejiang Chinese Medical University, Tiyuchang Rd 453, Hangzhou 310007, People's Republic of China
| | - Yayu Li
- Hangzhou Hospital of Traditional Chinese Medicine, Hangzhou TCM Hospital Affiliated to Zhejiang Chinese Medical University, Tiyuchang Rd 453, Hangzhou 310007, People's Republic of China
| | - Fenggui Zhu
- Hangzhou Hospital of Traditional Chinese Medicine, Hangzhou TCM Hospital Affiliated to Zhejiang Chinese Medical University, Tiyuchang Rd 453, Hangzhou 310007, People's Republic of China
| | - Bo Xie
- Hangzhou Hospital of Traditional Chinese Medicine, Hangzhou TCM Hospital Affiliated to Zhejiang Chinese Medical University, Tiyuchang Rd 453, Hangzhou 310007, People's Republic of China; Hangzhou Third People's Hospital, Hangzhou Third Hospital Affiliated to Zhejiang Chinese Medical University, Affiliated Hangzhou Dermatology Hospital, Zhejiang University School of Medicine, Westlake Ave 38, Hangzhou 310009, People's Republic of China.
| |
Collapse
|
34
|
Lipoprotein (a) is related to In-Stent neoatherosclerosis incidence rate and plaque vulnerability: Optical Coherence Tomography Study. Int J Cardiovasc Imaging 2023; 39:275-284. [PMID: 36315364 PMCID: PMC9870965 DOI: 10.1007/s10554-022-02736-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Accepted: 09/24/2022] [Indexed: 11/05/2022]
Abstract
BACKGROUND In-stent neoatherosclerosis (ISNA) is an important reason for stent failure. High lipoprotein (a) [Lp (a)] level is an independent predictor of in-stent restenosis (ISR). To date, the relationship between the level of serum Lp (a) and the incidence rate and vulnerability of ISNA has never been verified. METHODS A total of 119 patients with 125 drug-eluting stent ISR lesions who underwent percutaneous coronary intervention guided by optical coherence tomography were enrolled in this study. According to their Lp (a) level, the patients were divided into two groups [high Lp (a) group ≥ 30 mg/dL, n = 47; or low Lp (a) group < 30 mg/dL, n = 72]. The clinical baseline, angiographic characteristics, and optical coherence tomography data of both groups were recorded and analyzed. RESULTS No significant differences in clinical and angiographic characteristics were found between the two groups (P > 0.05). The incidence rate of ISNA in the high Lp (a) group was significantly higher than that in the low Lp (a) group (94.0% [n = 47] vs. 52.0% [n = 39], P < 0.001). The incidence rate of thin-cap fibroatheroma in ISR lesions was significantly higher in the high Lp (a) group than in the low Lp (a) group (42% [n = 21] vs. 5.3% [n = 4], P < 0.001). CONCLUSION A high Lp (a) level is associated with the high incidence rate and plaque vulnerability of ISNA.
Collapse
|
35
|
Grundler F, Viallon M, Mesnage R, Ruscica M, von Schacky C, Madeo F, Hofer SJ, Mitchell SJ, Croisille P, Wilhelmi de Toledo F. Long-term fasting: Multi-system adaptations in humans (GENESIS) study-A single-arm interventional trial. Front Nutr 2022; 9:951000. [PMID: 36466423 PMCID: PMC9713250 DOI: 10.3389/fnut.2022.951000] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Accepted: 10/31/2022] [Indexed: 09/10/2024] Open
Abstract
Fasting provokes fundamental changes in the activation of metabolic and signaling pathways leading to longer and healthier lifespans in animal models. Although the involvement of different metabolites in fueling human fasting metabolism is well known, the contribution of tissues and organs to their supply remains partly unclear. Also, changes in organ volume and composition remain relatively unexplored. Thus, processes involved in remodeling tissues during fasting and food reintroduction need to be better understood. Therefore, this study will apply state-of-the-art techniques to investigate the effects of long-term fasting (LF) and food reintroduction in humans by a multi-systemic approach focusing on changes in body composition, organ and tissue volume, lipid transport and storage, sources of protein utilization, blood metabolites, and gut microbiome profiles in a single cohort. This is a prospective, single-arm, monocentric trial. One hundred subjects will be recruited and undergo 9 ± 3 day-long fasting periods (250 kcal/day). We will assess changes in the composition of organs, bones and blood lipid profiles before and after fasting, as well as high-density lipoprotein (HDL) transport and storage, untargeted metabolomics of peripheral blood mononuclear cells (PBMCs), protein persulfidation and shotgun metagenomics of the gut microbiome. The first 32 subjects, fasting for 12 days, will be examined in more detail by magnetic resonance imaging (MRI) and spectroscopy to provide quantitative information on changes in organ volume and function, followed by an additional follow-up examination after 1 and 4 months. The study protocol was approved by the ethics board of the State Medical Chamber of Baden-Württemberg on 26.07.2021 and registered at ClinicalTrials.gov (NCT05031598). The results will be disseminated through peer-reviewed publications, international conferences and social media. Clinical trial registration [ClinicalTrials.gov], identifier [NCT05031598].
Collapse
Affiliation(s)
| | - Magalie Viallon
- UJM-Saint-Etienne, INSA, CNRS UMR 5520, INSERM U1206, CREATIS, F-42023, Université de Lyon, Saint-Étienne, France
- Department of Radiology, University Hospital Saint-Étienne, Saint-Étienne, France
| | - Robin Mesnage
- Buchinger Wilhelmi Clinic, Überlingen, Germany
- Department of Medical and Molecular Genetics, Faculty of Life Sciences and Medicine, King’s College London, London, United Kingdom
| | - Massimiliano Ruscica
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milan, Italy
| | | | - Frank Madeo
- Institute of Molecular Biosciences, NAWI Graz, University of Graz, Graz, Austria
- BioHealth Graz, Graz, Austria
- BioTechMed Graz, Graz, Austria
| | - Sebastian J. Hofer
- Institute of Molecular Biosciences, NAWI Graz, University of Graz, Graz, Austria
- BioHealth Graz, Graz, Austria
- BioTechMed Graz, Graz, Austria
| | - Sarah J. Mitchell
- Department of Health Sciences and Technology, ETH Zürich, Schwerzenbach, Switzerland
| | - Pierre Croisille
- UJM-Saint-Etienne, INSA, CNRS UMR 5520, INSERM U1206, CREATIS, F-42023, Université de Lyon, Saint-Étienne, France
- Department of Radiology, University Hospital Saint-Étienne, Saint-Étienne, France
| | | |
Collapse
|
36
|
Ruscica M, Rizzuto AS, Corsini A. Role of lipoprotein(a) in plaque progression. Eur Heart J Suppl 2022; 24:I72-I75. [PMID: 36380804 PMCID: PMC9653156 DOI: 10.1093/eurheartjsupp/suac071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Identified by Berg in 1963, lipoprotein(a) represents a key contemporary residual risk pathway in atherosclerotic cardiovascular disease (ASCVD) secondary prevention. Indeed, epidemiological and genetic studies have undoubtedly demonstrated that lipoprotein(a) is one of the strongest causal risk factors of ASCVD. Although a risk threshold has been set between 30 and 50 mg/dL, depending on the ethnicity, a linear risk gradient across the distribution has been demonstrated. In the context of the atherosclerotic process, hyperlipoproteinaemia(a) contributes to the atherosclerotic plaque formation by deposition of cholesterol in the same manner as low-density lipoprotein (LDL) cholesterol, due to the LDL particle component of lipoprotein(a). Lipoprotein(a) accumulates in human coronary and carotid atherosclerotic lesions. High concentrations of lipoprotein(a) are associated with accelerated progression of the necrotic core, but not with coronary calcium score (CAC), although in the latter case, the evaluation of lipoprotein(a) can overcome the potential limitation of CAC to capture the totality of ASCVD risk in asymptomatic individuals. Finally, in the absence of a pharmacological approach to lower lipoprotein(a) to the extent required to achieve a cardiovascular benefit, implementation strategies that increase awareness among the population, patients, and healthcare providers on the importance of lipoprotein(a) in the development of ASCVD are eagerly needed.
Collapse
Affiliation(s)
| | | | - Alberto Corsini
- Department of Pharmacological and Biomolecular Sciences, University of Milan
| |
Collapse
|
37
|
Li JJ, Ma CS, Zhao D, Yan XW. Lipoprotein(a) and Cardiovascular Disease in Chinese Population: A Beijing Heart Society Expert Scientific Statement. JACC. ASIA 2022; 2:653-665. [PMID: 36444328 PMCID: PMC9700018 DOI: 10.1016/j.jacasi.2022.08.015] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Revised: 08/12/2022] [Accepted: 08/23/2022] [Indexed: 11/17/2022]
Abstract
Elevated concentration of lipoprotein(a) [Lp(a)] is an independent risk factor for atherosclerotic cardiovascular disease, including coronary artery disease, stroke, peripheral artery disease, and so on. Emerging data suggest that Lp(a) contributes to the increased risk for cardiovascular events even in the setting of effective reduction of plasma low-density lipoprotein cholesterol. Nevertheless, puzzling issues exist covering potential genetic factors, Lp(a) assay, possible individuals for analysis, a cutoff point of increased risk, and clinical interventions. In the Chinese population, Lp(a) exhibited a distinctive prevalence and regulated various cardiovascular diseases in specific ways. Hence, it is valuable to clarify the role of Lp(a) in cardiovascular diseases and explore prevention and control measures for the increase in Lp(a) prevalence in the Chinese population. This Beijing Heart Society experts' scientific statement will present the detailed knowledge concerning Lp(a)-related studies combined with Chinese population observations to provide the key points of reference.
Collapse
Key Words
- AMI, acute myocardial infarction
- ASCVD, atherosclerotic cardiovascular disease
- Apo, apolipoprotein
- CAD, coronary artery disease
- CAVS, calcific aortic valve stenosis
- CVD, cardiovascular disease
- CVE, cardiovascular event
- FH, familial hypercholesterolemia
- GWAS, genome-wide association analysis
- KIV, Kringle IV
- LA, lipoprotein apheresis
- LDL-C, low-density lipoprotein cholesterol
- Lp(a), lipoprotein(a)
- MACE, major adverse cardiovascular events
- OxPL, oxidized phospholipids
- PCSK9, proprotein convertase subtilisin/kexin type 9
- SNP, single nucleotide polymorphism
- T2DM, type 2 diabetes mellitus
- atherosclerotic cardiovascular disease
- calcific aortic value stenosis
- lipoprotein(a)
- scientific statement
Collapse
Affiliation(s)
- Jian-Jun Li
- Fuwai Hospital, Chinese Academy of Medical Sciences, Beijing, China
| | - Chang-Sheng Ma
- Beijing Anzhen Hospital Affiliated to Capital Medical University, Beijing, China
| | - Dong Zhao
- Beijing Anzhen Hospital Affiliated to Capital Medical University, Beijing, China
| | - Xiao-Wei Yan
- Beijing Union Medical College Hospital, Beijing, China
| |
Collapse
|
38
|
Lipoprotein a Combined with Fibrinogen as an Independent Predictor of Long-Term Prognosis in Patients with Acute Coronary Syndrome: A Multi-Center Retrospective Study. J Cardiovasc Dev Dis 2022; 9:jcdd9100322. [PMID: 36286274 PMCID: PMC9604333 DOI: 10.3390/jcdd9100322] [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: 08/05/2022] [Revised: 09/15/2022] [Accepted: 09/20/2022] [Indexed: 11/20/2022] Open
Abstract
Background: Patients with acute coronary syndrome (ACS) still have a high risk of recurrence of major adverse cardiovascular and cerebrovascular events (MACCE). However, there are rare studies on the prediction of MACCE in patients with ACS using lipoprotein a [Lp(a)] combined with fibrinogen. The aim of this study was to analyze the predictive value of Lp(a) combined with fibrinogen for the long-term prognosis of patients with ACS. Methods: 804 patients with ACS admitted to 11 tertiary general hospitals in Chengdu from January 2017 to June 2019 were included in the study. According to the Lp(a) 300 mg/L, patients were assigned to the non-high Lp(a) group and high Lp(a) group. Patients were assigned to the non-high or high fibrinogen groups using the fibrinogen level of 3.08 g/L. Subsequently, patients were divided into group A, B, or C by Lp(a) combined with fibrinogen. The study endpoints were MACCE, including all-cause death, non-fatal myocardial infarction, non-fatal stroke, and revascularization. The incidences of MACCE among groups were compared. Lp(a), fibrinogen, Lp(a) combined with fibrinogen classifications were each added into the basic model to construct three new models. The C-index, net reclassification index (NRI) and integrated discrimination improvement (IDI) of the three new models were then compared. Results: The median follow-up was 16 months. During follow-up, the cumulative incidence of MACCE in group C was significantly higher than that measured in group A and B (p < 0.001). The results of the multivariate Cox regression analysis of MACCE showed that Lp(a) ≥300 mg/L with fibrinogen ≥3.08 g/L was an independent predictor of MACCE. According to the GRACE score and the statistical analyses, the basic model was constructed, which had a C-index of 0.694. The C-index, NRI, and IDI of the new model constructed using the basic model + Lp(a) combined with fibrinogen classification were 0.736, 0.095, and 0.094 respectively. Conclusions: Single Lp(a), single fibrinogen and Lp(a) combined with fibrinogen were independent predictors of MACCE in patients with ACS. The predictive value of Lp(a) combined with fibrinogen in patients with ACS was better than that of single Lp(a) and single fibrinogen.
Collapse
|
39
|
Jia X, Al Rifai M, Saeed A, Ballantyne CM, Virani SS. PCSK9 Inhibitors in the Management of Cardiovascular Risk: A Practical Guidance. Vasc Health Risk Manag 2022; 18:555-566. [PMID: 35898405 PMCID: PMC9309324 DOI: 10.2147/vhrm.s275739] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Accepted: 07/13/2022] [Indexed: 01/19/2023] Open
Abstract
Proprotein convertase subtilisin/kexin type 9 (PCSK9) inhibitors are potent medications in the toolkit for treatment of atherosclerotic cardiovascular disease. These agents have been well studied in clinical trials supporting their efficacy in dramatically reducing low-density lipoprotein cholesterol (LDL-C) and impact on cardiovascular outcomes. Since the approval of commercial use for PCSK9 inhibitors in 2015, we have also gained significant experience in the use of these therapeutics in the real-world setting. In this article, we review current guideline recommendations, clinical trial evidence on efficacy and safety as well as data on cost-effectiveness, prescription and adherence. We focus primarily on the monoclonal antibody class of PCSK9 inhibitors in this review while also touching on other types of therapeutics that are under development.
Collapse
Affiliation(s)
- Xiaoming Jia
- Department of Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Mahmoud Al Rifai
- Department of Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Anum Saeed
- Department of Medicine, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | | | - Salim S Virani
- Department of Medicine, Baylor College of Medicine, Houston, TX, USA.,Department of Medicine, Michael E. DeBakey Veterans Affairs Medical Center, Houston, TX, USA
| |
Collapse
|
40
|
Ruscica M, Sirtori CR, Carugo S, Calder PC, Corsini A. OMEGA-3 AND CARDIOVASCULAR PREVENTION – IS THIS STILL A CHOICE? Pharmacol Res 2022; 182:106342. [DOI: 10.1016/j.phrs.2022.106342] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Revised: 07/01/2022] [Accepted: 07/01/2022] [Indexed: 02/07/2023]
|
41
|
Zhou L, Liu J, An Y, Wang Y, Wang G. Plasma Homocysteine Level Is Independently Associated With Conventional Atherogenic Lipid Profile and Remnant Cholesterol in Adults. Front Cardiovasc Med 2022; 9:898305. [PMID: 35770226 PMCID: PMC9234129 DOI: 10.3389/fcvm.2022.898305] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Accepted: 05/10/2022] [Indexed: 11/13/2022] Open
Abstract
BackgroundHomocysteine (Hcy) is an independent risk factor for cardiovascular disease, while mechanisms are unclear. Despite inconsistent and limited, epidemiological and experimental studies indicated that hyperhomocysteinemia (HHcy) affected lipid metabolism. This study aims to investigate the association of plasma Hcy with traditional lipid profiles and remnant cholesterol (RC) in Chinese adults.MethodsIn total, 7,898 subjects aged 20–79 years who underwent a physical examination at Beijing Chao-Yang Hospital in Beijing were included in this study. Fasting plasma total cholesterol (TC), triglyceride (TG), high-density lipoprotein cholesterol (HDL-C), low-density lipoprotein cholesterol (LDL-C), apolipoprotein A1 (ApoA1), apolipoprotein B (ApoB), lipoprotein (a) [Lp(a)], Hcy, and other metabolic risk factors were measured by routine automated laboratory methods. RC was calculated as TC minus HDL-C and LDL-C. The linear regression model and logistic regression model were used to assess the relationship between Hcy and lipids after adjusting potential confounders.ResultsOf the subjects, the median level of plasma Hcy was 13.0 μmol/L and 32.3% had HHcy. Plasma Hcy was negatively associated with HDL-C, ApoA1, and Lp(a) and positively associated with TG levels after adjusting age, sex, body mass index, blood pressure, alanine transaminase, aspartate transaminase, creatinine, uric acid, and glucose. HHcy significantly increased the risk of low HDL-C [odds ratio (OR) 1.26; 95%CI (1.11–1.44); p < 0.001]. The net mediation effects of ApoA1 on the relationship between Hcy and HDL-C before and after adjusting confounders were 46.9 and 30.6%, respectively. More interestingly, the RC level was significantly elevated in subjects with HHcy after adjusting other influencing factors (p = 0.025). Hcy presented a positive correlation with RC levels after adjusting the above confounding factors (β = 0.073, p = 0.004), and the correlation was still significant even after controlling other lipids, including TG, LDL-C, HDL-C, ApoA1, ApoB, and Lp(a).ConclusionOur study showed that plasma Hcy was not only significantly associated with conventional atherogenic lipids but also independently correlated with RC levels beyond other lipids after controlling potential confounders. This finding proposes that identifying Hcy-related dyslipidemia risk, both traditional lipids and RC residual risk, is clinically relevant as we usher in a new era of targeting Hcy-lowering therapies to fight against dyslipidemia or even cardiovascular disease.
Collapse
Affiliation(s)
- Liyuan Zhou
- Department of Endocrinology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Jia Liu
- Department of Endocrinology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Yu An
- Department of Endocrinology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Ying Wang
- Medical Examination Center, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
- *Correspondence: Ying Wang,
| | - Guang Wang
- Department of Endocrinology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
- Guang Wang,
| |
Collapse
|
42
|
Cremonini AL, Pasta A, Carbone F, Visconti L, Casula M, Elia E, Bonaventura A, Liberale L, Bertolotto M, Artom N, Minetti S, Contini P, Verzola D, Pontremoli R, Viazzi F, Viviani GL, Bertolini S, Pende A, Montecucco F, Pisciotta L. Lipoprotein(a) Modulates Carotid Atherosclerosis in Metabolic Syndrome. Front Mol Biosci 2022; 9:854624. [PMID: 35755826 PMCID: PMC9214261 DOI: 10.3389/fmolb.2022.854624] [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: 01/14/2022] [Accepted: 05/09/2022] [Indexed: 11/15/2022] Open
Abstract
Background and Aim: High lipoprotein(a) [Lp(a)] is a well-established cardiovascular (CV) risk factor, but the effect of mildly elevated Lp(a) on CV health is largely unknown. Our aim was to evaluate if Lp(a) is associated with the severity of carotid atherosclerosis (CA) in the specific subset of metabolic syndrome (MetS). Patients and Methods: Subjects with diagnosed MetS and ultrasound-assessed CA were enrolled. Those patients were categorized according to the severity of CA (moderate vs. severe), and the circulating levels of Lp(a) alongside with clinical, anthropometric, and biochemical data were collected. Results: Sixty-five patients were finally included: twenty-five with moderate and forty with severe CA (all with asymptomatic disease). Intergroup comparison showed Lp(a) as the only significantly different variable [6 (2-12) mg/dl vs. 11.5 (6-29.5) mg/dl; p = 0.018]. Circulating levels of Lp(a) were also confirmed as the only variable independently associated with severity of CA at logistic regression analysis [OR 2.9 (95% CI 1.1-7.8); p = 0.040]. ROC curve analysis for Lp(a) confirmed a serum level of 10 mg/dl as the best cut-off value [AUC 0.675 (95% CI 0.548-0.786)]. Although sensitivity and specificity were suboptimal (69.0 and 70.4%, respectively)-likely due to the small sample size-this result is in line with those previously reported in the literature. Conclusion: Lp(a) is independently associated with severity of CA in the subgroup of MetS patients.
Collapse
Affiliation(s)
- Anna Laura Cremonini
- Department of Internal Medicine, University of Genoa, Genoa, Italy
- IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Andrea Pasta
- Department of Internal Medicine, University of Genoa, Genoa, Italy
| | - Federico Carbone
- Department of Internal Medicine, University of Genoa, Genoa, Italy
- IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Luca Visconti
- Department of Internal Medicine, University of Genoa, Genoa, Italy
| | - Matteo Casula
- Department of Internal Medicine, University of Genoa, Genoa, Italy
| | - Edoardo Elia
- Department of Internal Medicine, University of Genoa, Genoa, Italy
| | - Aldo Bonaventura
- Department of Internal Medicine, University of Genoa, Genoa, Italy
- Internal Medicine Department Ospedale di Circolo e Fondazione Macchi, ASST Sette Laghi, Varese, Italy
| | - Luca Liberale
- Department of Internal Medicine, University of Genoa, Genoa, Italy
- IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Maria Bertolotto
- Department of Internal Medicine, University of Genoa, Genoa, Italy
| | - Nathan Artom
- Department of Internal Medicine, Ospedale S. Paolo di Savona, Savona, Italy
| | - Silvia Minetti
- Department of Internal Medicine, University of Genoa, Genoa, Italy
| | - Paola Contini
- Department of Internal Medicine, University of Genoa, Genoa, Italy
- IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Daniela Verzola
- Department of Internal Medicine, University of Genoa, Genoa, Italy
| | - Roberto Pontremoli
- Department of Internal Medicine, University of Genoa, Genoa, Italy
- IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Francesca Viazzi
- Department of Internal Medicine, University of Genoa, Genoa, Italy
- IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | | | | | - Aldo Pende
- Department of Internal Medicine, University of Genoa, Genoa, Italy
- IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Fabrizio Montecucco
- Department of Internal Medicine, University of Genoa, Genoa, Italy
- IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Livia Pisciotta
- Department of Internal Medicine, University of Genoa, Genoa, Italy
- IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| |
Collapse
|
43
|
Gao F, Li YP, Ma XT, Wang ZJ, Shi DM, Zhou YJ. Effect of Alirocumab on Coronary Calcification in Patients With Coronary Artery Disease. Front Cardiovasc Med 2022; 9:907662. [PMID: 35600486 PMCID: PMC9120536 DOI: 10.3389/fcvm.2022.907662] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Accepted: 04/19/2022] [Indexed: 11/25/2022] Open
Abstract
Background Proprotein convertase subtilisin kexin type 9 (PCSK9) inhibitors have been documented with significantly reduction in LDL cholesterol levels and cardiovascular events. However, evidence regarding the impact of PCSK9 inhibitors on coronary calcification is limited. Methods Eligible patients with intermediate coronary lesions and elevated LDL cholesterol values were randomized to either alirocumab 75 mg Q2W plus statin (atorvastatin 20 mg/day or rosuvastatin 10 mg/day) therapy or standard statin therapy. Calcium score based on coronary computed tomographic angiography at baseline and follow up were compared. Results Compared with baseline levels, LDL cholesterol were significantly decreased in both groups, while the absolute reduction of LDL cholesterol levels were higher in patients treated with alirocumab (1.69 ± 0.52 vs. 0.92 ± 0.60, P < 0.0001). Additionally, patients in alirocumab group demonstrated a significant reduction of Lp(a) levels, whereas it was not observed in the standard statin group. Notably, greater increases in the percentage changes of CAC score (10.6% [6.3–23.3] vs. 2.9% [−6.7–8.3]; P < 0.0001) were observed in the statin group compared to the alirocumab group. Consistently, CAC progression was significantly lower in the alirocumab group than in the standard statin group (0.6 ± 2.2% vs. 2.7 ± 2.3%; P = 0.002). Conclusions Study indicated that administration of the PCSK9 inhibitors to statins produced significantly lower rate of CAC progression in patients with coronary artery disease. Further studies with CAC progression and their clinical outcomes are needed. Trial Registration ClinicalTrials.gov, Identifier: NCT04851769.
Collapse
|
44
|
Waldmann E, Wu L, Busygina K, Altenhofer J, Henze K, Folwaczny A, Parhofer KG. Effect of PCSK9 inhibition with evolocumab on lipoprotein subfractions in familial dysbetalipoproteinemia (type III hyperlipidemia). PLoS One 2022; 17:e0265838. [PMID: 35320320 PMCID: PMC8942248 DOI: 10.1371/journal.pone.0265838] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Accepted: 03/08/2022] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND AND AIMS Familial dysbetalipoproteinemia (FDBL) is a rare inborn lipid disorder characterized by the formation of abnormal triglyceride- and cholesterol-rich lipoproteins (remnant particles). Patients with FDBL have a high risk for atherosclerotic disease. The effect of PCSK9 inhibition on lipoproteins and its subfractions has not been evaluated in FDBL. METHODS Three patients (65±7 years, 23±3 kg/m2, 2 females) with FDBL (diagnosed by isoelectrofocusing) and atherosclerosis (coronary and/or cerebro-vascular and/or peripheral arterial disease) resistant or intolerant to statin and fibrate therapy received evolocumab (140mg every 14 days). In addition to a fasting lipid profile (preparative ultracentrifugation), apoB and cholesterol concentrations were determined in 15 lipoprotein-subfractions (density gradient ultracentrifugation; d 1.006-1.21g/ml) before and after 12 weeks of evolocumab treatment. Patients with LDL-hypercholesterolemia (n = 8, 56±8 years, 31±7 kg/m2) and mixed hyperlipidemia (n = 5, 68±12 years, 30±1 kg/m2) also receiving evolocumab for 12 weeks were used for comparison. RESULTS All patients tolerated PCSK9 inhibition well. PCSK9 inhibitors reduced cholesterol (29-37%), non-HDL-cholesterol (36-50%) and apoB (40-52%) in all patient groups including FDBL. In FDBL, PCSK9 inhibition reduced VLDL-cholesterol and the concentration of apoB containing lipoproteins throughout the whole density spectrum (VLDL, IDL, remnants, LDL). Lipoprotein(a) was decreased in all patient groups to a similar extent. CONCLUSIONS This indicates that the dominant fraction of apoB-containing lipoproteins is reduced with PCSK9 inhibition, i.e. LDL in hypercholesterolemia and mixed hyperlipidemia, and cholesterol-rich VLDL, remnants and LDL in FDBL. PCSK9 inhibition may be a treatment option in patients with FDBL resistant or intolerant to statin and/or fibrate therapy.
Collapse
Affiliation(s)
- Elisa Waldmann
- Medical Department IV, LMU Klinikum Grosshadern, Munich, Germany
| | - Liya Wu
- Medical Department IV, LMU Klinikum Grosshadern, Munich, Germany
| | | | - Julia Altenhofer
- Medical Department IV, LMU Klinikum Grosshadern, Munich, Germany
| | - Kerstin Henze
- Medical Department IV, LMU Klinikum Grosshadern, Munich, Germany
| | | | - Klaus G. Parhofer
- Medical Department IV, LMU Klinikum Grosshadern, Munich, Germany
- * E-mail:
| |
Collapse
|
45
|
Sirtori CR, Corsini A, Ruscica M. The Role of High-Density Lipoprotein Cholesterol in 2022. Curr Atheroscler Rep 2022; 24:365-377. [PMID: 35274229 PMCID: PMC8913032 DOI: 10.1007/s11883-022-01012-y] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/31/2022] [Indexed: 12/23/2022]
Abstract
PURPOSE OF THE REVIEW High-density lipoproteins (HDL) are responsible for the transport in plasma of a large fraction of circulating lipids, in part from tissue mobilization. The evaluation of HDL-associated cholesterol (HDL-C) has provided a standard method for assessing cardiovascular (CV) risk, as supported by many contributions on the mechanism of this arterial benefit. The present review article will attempt to investigate novel findings on the role and mechanism of HDL in CV risk determination. RECENT FINDINGS The most recent research has been aimed to the understanding of how a raised functional capacity of HDL, rather than elevated levels per se, may be responsible for the postulated CV protection. Markedly elevated HDL-C levels appear instead to be associated to a raised coronary risk, indicative of a U-shaped relationship. While HDL-C reduction is definitely related to a raised CV risk, HDL-C elevations may be linked to non-vascular diseases, such as age-related macular disease. The description of anti-inflammatory, anti-oxidative and anti-infectious properties has indicated potential newer areas for diagnostic and therapeutic approaches. In the last two decades inconclusive data have arisen from clinical trials attempting to increase HDL-C pharmacologically or by way of recombinant protein infusions (most frequently with the mutant A-I Milano); prevention of stent occlusion or heart failure treatment have shown instead significant promise. Targeted clinical studies are still ongoing.
Collapse
Affiliation(s)
- Cesare R Sirtori
- Department of Pharmacological and Biomolecular Sciences, Università Degli Studi Di Milano, Milan, Italy.
| | - Alberto Corsini
- Department of Pharmacological and Biomolecular Sciences, Università Degli Studi Di Milano, Milan, Italy
| | - Massimiliano Ruscica
- Department of Pharmacological and Biomolecular Sciences, Università Degli Studi Di Milano, Milan, Italy.
| |
Collapse
|
46
|
Blom DJ, Marais AD, Moodley R, van der Merwe N, van Tonder A, Raal FJ. RNA-based therapy in the management of lipid disorders: a review. Lipids Health Dis 2022; 21:41. [PMID: 35459248 PMCID: PMC9034497 DOI: 10.1186/s12944-022-01649-3] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Accepted: 03/31/2022] [Indexed: 11/10/2022] Open
Abstract
This review focuses on antisense oligonucleotides and small interfering ribonucleic acid therapies approved or under development for the management of lipid disorders. Recent advances in RNA-based therapeutics allow tissue-specific targeting improving safety. Multiple potential target proteins have been identified and RNA-based therapeutics have the potential to significantly improve outcomes for patients with or at risk for atherosclerotic cardiovascular disease. The advantages of RNA-based lipid modifying therapies include the ability to reduce the concentration of almost any target protein highly selectively, allowing for more precise control of metabolic pathways than can often be achieved with small molecule-based drugs. RNA-based lipid modifying therapies also make it possible to reduce the expression of target proteins for which there are no small molecule inhibitors. RNA-based therapies can also reduce pill burden as their administration schedule typically varies from weekly to twice yearly injections. The safety profile of most current RNA-based lipid therapies is acceptable but adverse events associated with various therapies targeting lipid pathways have included injection site reactions, inflammatory reactions, hepatic steatosis and thrombocytopenia. While the body of evidence for these therapies is expanding, clinical experience with these therapies is currently limited in duration and the results of long-term studies are eagerly awaited.
Collapse
Affiliation(s)
- Dirk Jacobus Blom
- Department of Medicine, Division of Lipidology and Hatter Institute for Cardiovascular Research in Africa, University of Cape Town, Cape Town, South Africa.
| | - Adrian David Marais
- Division of Chemical Pathology, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Rajen Moodley
- Netcare Umhlanga Medical Center, Umhlanga, KwaZulu Natal, South Africa
| | | | | | - Frederick Johan Raal
- Carbohydrate and Lipid Metabolism Research Unit, Faculty of Health Sciences, University of Witwatersrand, Johannesburg, South Africa
| |
Collapse
|
47
|
Koutsogianni AD, Liberopoulos E, Tellis K, Tselepis AD. Oxidized phospholipids and lipoprotein(a): An update. Eur J Clin Invest 2022; 52:e13710. [PMID: 34837383 DOI: 10.1111/eci.13710] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2021] [Revised: 10/21/2021] [Accepted: 10/21/2021] [Indexed: 12/17/2022]
Abstract
Over the past few years, there has been an undiminished interest in lipoprotein(a) [Lp(a)] and oxidized phospholipids (OxPLs), mainly carried on this lipoprotein. Elevated Lp(a) has been established as an independent causal risk factor for cardiovascular disease. OxPLs play an important role in atherosclerosis. The main questions that remain to be answered, however, is to what extent OxPLs contribute to the atherogenicity of Lp(a), what effect hypolipidaemic medications may have on their levels and the potential clinical benefit of their reduction. This narrative review aimed to summarize currently available data on OxPLs and cardiovascular risk, as well as the effect of established and emerging hypolipidaemic medications on Lp(a)-OxPLs.
Collapse
Affiliation(s)
| | - Evangelos Liberopoulos
- Department of Internal Medicine, Faculty of Medicine, University of Ioannina, Ioannina, Greece
| | - Konstantinos Tellis
- Department of Chemistry, Atherothrombosis Research Centre/Laboratory of Biochemistry, University of Ioannina, Ioannina, Greece
| | - Alexandros D Tselepis
- Department of Chemistry, Atherothrombosis Research Centre/Laboratory of Biochemistry, University of Ioannina, Ioannina, Greece
| |
Collapse
|
48
|
Lipoprotein(a) Serum Levels Predict Pulse Wave Velocity in Subjects in Primary Prevention for Cardiovascular Disease with Large Apo(a) Isoforms: Data from the Brisighella Heart Study. Biomedicines 2022; 10:biomedicines10030656. [PMID: 35327457 PMCID: PMC8945189 DOI: 10.3390/biomedicines10030656] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Revised: 03/04/2022] [Accepted: 03/09/2022] [Indexed: 11/17/2022] Open
Abstract
In the last decades, high serum levels of lipoprotein(a) (Lp(a)) have been associated with increased cardiovascular disease (CVD) risk, in particular among individuals with smaller apolipoprotein(a) (apo(a)) isoforms than those with larger sizes. The aim of our analysis was to evaluate whether Lp(a) levels could predict early vascular aging, and whether smaller apo(a) isoforms had a predictive value for vascular aging different than larger apo(a) isoforms in a cohort of subjects free from CVD. We considered the data of a subset of Brisighella Heart Study (BHS) participants free from CVD (462 men and 516 women) who were clinically evaluated during the 2012 BHS population survey. Predictors of arterial stiffness, measured as carotid-femoral pulse wave velocity (cfPWV) were estimated by the application of a step-wise linear regression model. In our cohort, there were 511 subjects with small apo(a) size and 467 subjects with large apo(a) isoforms. Subjects with larger apo(a) isoform sizes had significantly lower serum levels of Lp(a). In the BHS subpopulation sample, cfPWV was predicted by age, systolic blood pressure (SBP), serum levels of high-density lipoprotein cholesterol (HDL-C), triglycerides (TG) and sex, higher HDL-C serum levels and female sex associated with lower values of cfPWV. In subjects with smaller apo(a) isoform sizes, predictors of cfPWV were age, SBP, sex and serum levels of HDL-C, being higher HDL-C serum levels and female sex associated to lower values of cfPWV. In subjects with larger apo(a) isoform sizes, cfPWV was predicted by age, SBP, serum levels of Lp(a) and sex, with female sex associated with lower values of cfPWV. In our subpopulation sample, Lp(a) did not predict cfPWV. However, in subjects with large apo(a) isoform sizes, Lp(a) was a significant predictor of arterial stiffness.
Collapse
|
49
|
Meroni M, Longo M, Lombardi R, Paolini E, Macchi C, Corsini A, Sirtori CR, Fracanzani AL, Ruscica M, Dongiovanni P. Low Lipoprotein(a) Levels Predict Hepatic Fibrosis in Patients With Nonalcoholic Fatty Liver Disease. Hepatol Commun 2022; 6:535-549. [PMID: 34677008 PMCID: PMC8870034 DOI: 10.1002/hep4.1830] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Revised: 08/30/2021] [Accepted: 09/08/2021] [Indexed: 12/21/2022] Open
Abstract
Dyslipidemia and cardiovascular complications are comorbidities of nonalcoholic fatty liver disease (NAFLD), which ranges from simple steatosis to nonalcoholic steatohepatitis, fibrosis, and cirrhosis up to hepatocellular carcinoma. Lipoprotein(a) (Lp(a)) has been associated with cardiovascular risk and metabolic abnormalities, but its impact on the severity of liver damage in patients with NAFLD remains to be clarified. Circulating Lp(a) levels were assessed in 600 patients with biopsy-proven NAFLD. The association of Lp(a) with liver damage was explored by categorizing serum Lp(a) into quartiles. The receiver operating characteristic curve was used to analyze the accuracy of serum Lp(a) in hepatic fibrosis prediction. Hepatic expression of lipoprotein A (LPA) and of genes involved in lipid metabolism and fibrogenic processes were evaluated by RNA sequencing in a subset of patients with NAFLD for whom Lp(a) dosage was available (n = 183). In patients with NAFLD, elevated Lp(a) levels were modestly associated with circulating lipids, carotid plaques, and hypertension (P < 0.05). Conversely, patients with low serum Lp(a) displayed insulin resistance (P < 0.05), transaminase elevation (P < 0.05), and increased risk of developing severe fibrosis (P = 0.007) and cirrhosis (P = 0.002). In addition, the diagnostic accuracy of Lp(a) in predicting fibrosis increased by combining it with transaminases (area under the curve fibrosis stage 4, 0.87; P < 0.0001). Hepatic LPA expression reflected serum Lp(a) levels (P = 0.018), and both were reduced with the progression of NAFLD (P < 0.05). Hepatic LPA messenger RNA levels correlated with those of genes involved in lipoprotein release, lipid synthesis, and fibrogenesis (P < 0.05). Finally, transmembrane 6 superfamily member 2 (TM6SF2) rs58542926, apolipoprotein E (ApoE) rs445925, and proprotein convertase subtilisin/kexin type 9 (PCSK9) rs7552841, known variants that modulate circulating lipids, may influence serum Lp(a) levels (P < 0.05). Conclusion: Circulating Lp(a) combined with transaminases may represent a novel noninvasive biomarker to predict advanced fibrosis in patients with NAFLD.
Collapse
Affiliation(s)
- Marica Meroni
- General Medicine and Metabolic DiseasesFondazione Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Cà Granda Ospedale Maggiore PoliclinicoMilanItaly
| | - Miriam Longo
- General Medicine and Metabolic DiseasesFondazione Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Cà Granda Ospedale Maggiore PoliclinicoMilanItaly.,Department of Clinical Sciences and Community HealthUniversità degli Studi di MilanoMilanItaly
| | - Rosa Lombardi
- General Medicine and Metabolic DiseasesFondazione Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Cà Granda Ospedale Maggiore PoliclinicoMilanItaly.,Department of Pathophysiology and TransplantationUniversità degli Studi di MilanoMilanItaly
| | - Erika Paolini
- General Medicine and Metabolic DiseasesFondazione Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Cà Granda Ospedale Maggiore PoliclinicoMilanItaly.,Department of Pharmacological and Biomolecular SciencesUniversità degli Studi di MilanoMilanItaly
| | - Chiara Macchi
- Department of Pharmacological and Biomolecular SciencesUniversità degli Studi di MilanoMilanItaly
| | - Alberto Corsini
- Department of Pharmacological and Biomolecular SciencesUniversità degli Studi di MilanoMilanItaly.,Multimedica IRCCSSesto San GiovanniMilanItaly
| | - Cesare R Sirtori
- Department of Pharmacological and Biomolecular SciencesUniversità degli Studi di MilanoMilanItaly
| | - Anna Ludovica Fracanzani
- General Medicine and Metabolic DiseasesFondazione Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Cà Granda Ospedale Maggiore PoliclinicoMilanItaly.,Department of Pathophysiology and TransplantationUniversità degli Studi di MilanoMilanItaly
| | - Massimiliano Ruscica
- Department of Pharmacological and Biomolecular SciencesUniversità degli Studi di MilanoMilanItaly
| | - Paola Dongiovanni
- General Medicine and Metabolic DiseasesFondazione Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Cà Granda Ospedale Maggiore PoliclinicoMilanItaly
| |
Collapse
|
50
|
Xia J, Guo C, Cao H, Liu K, Peng W, Sun Y, Xie Y, Li B, Zhang F, Wen F, Zhang L. Impact of lipoprotein(a) level on cardiometabolic disease in the Chinese population: The CHCN-BTH Study. Eur J Clin Invest 2022; 52:e13689. [PMID: 34632581 DOI: 10.1111/eci.13689] [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] [Received: 08/05/2021] [Revised: 09/24/2021] [Accepted: 09/26/2021] [Indexed: 10/20/2022]
Abstract
BACKGROUND The emergence of promising compounds to lower lipoprotein(a) [Lp(a)] has increased the need for a precise characterisation and comparability assessment of Lp(a)-associated cardiometabolic disease risk. This study aimed to evaluate the distribution of Lp(a) levels in a Chinese population and characterise the association with cardiometabolic diseases. METHODS We assessed data from individuals from the Cohort Study on Chronic Diseases of the General Community Population in the Beijing-Tianjin-Hebei Region project. All Lp(a) measurements were performed in the same hospital. The cardiometabolic diseases considered were coronary heart disease (CHD), stroke, hypertension and type 2 diabetes (T2DM). RESULTS A total of 25343 individuals were included in the study. The median level of Lp(a) was 11.9 mg/dl (IQR 5.9 to 23.7 mg/dl), and higher Lp(a) levels showed a significant concentration-dependent association with CHD risk. Individuals with Lp(a) levels lower than the 25th percentile were at increased risk of hypertension (OR: 1.15, 95% CI: 1.06-1.25) and T2DM (OR: 1.15, 95% CI: 1.03-1.28); however, Lp(a) levels were not significantly associated with stroke. The addition of Lp(a) levels to the prognostic model led to a marginal but significant C-index, integrated discrimination improvement and net reclassification improvement. CONCLUSIONS In this large sample size study, we observed that elevated Lp(a) levels were significantly associated with CHD. Furthermore, we found that the lowest Lp(a) levels were also significantly associated with hypertension and T2DM. These results provide evidence for differential approaches to higher levels of Lp(a) in individuals with different cardiometabolic diseases.
Collapse
Affiliation(s)
- Juan Xia
- Department of Epidemiology and Health Statistics, School of Public Health, Capital Medical University, and Beijing Municipal Key Laboratory of Clinical Epidemiology, Beijing, China
| | - Chunyue Guo
- Department of Epidemiology and Health Statistics, School of Public Health, Capital Medical University, and Beijing Municipal Key Laboratory of Clinical Epidemiology, Beijing, China
| | - Han Cao
- Department of Epidemiology and Health Statistics, School of Public Health, Capital Medical University, and Beijing Municipal Key Laboratory of Clinical Epidemiology, Beijing, China
| | - Kuo Liu
- Department of Epidemiology and Health Statistics, School of Public Health, Capital Medical University, and Beijing Municipal Key Laboratory of Clinical Epidemiology, Beijing, China
| | - Wenjuan Peng
- Department of Epidemiology and Health Statistics, School of Public Health, Capital Medical University, and Beijing Municipal Key Laboratory of Clinical Epidemiology, Beijing, China
| | - Yanyan Sun
- Department of Epidemiology and Health Statistics, School of Public Health, Capital Medical University, and Beijing Municipal Key Laboratory of Clinical Epidemiology, Beijing, China
| | - Yunyi Xie
- Department of Epidemiology and Health Statistics, School of Public Health, Capital Medical University, and Beijing Municipal Key Laboratory of Clinical Epidemiology, Beijing, China
| | - Bingxiao Li
- Department of Epidemiology and Health Statistics, School of Public Health, Capital Medical University, and Beijing Municipal Key Laboratory of Clinical Epidemiology, Beijing, China
| | - Fengxu Zhang
- Department of Epidemiology and Health Statistics, School of Public Health, Capital Medical University, and Beijing Municipal Key Laboratory of Clinical Epidemiology, Beijing, China
| | - Fuyuan Wen
- Department of Epidemiology and Health Statistics, School of Public Health, Capital Medical University, and Beijing Municipal Key Laboratory of Clinical Epidemiology, Beijing, China
| | - Ling Zhang
- Department of Epidemiology and Health Statistics, School of Public Health, Capital Medical University, and Beijing Municipal Key Laboratory of Clinical Epidemiology, Beijing, China
| |
Collapse
|