1
|
Zarandi PK, Ghiasi M, Heiat M. The role and function of lncRNA in ageing-associated liver diseases. RNA Biol 2025; 22:1-8. [PMID: 39697114 PMCID: PMC11660375 DOI: 10.1080/15476286.2024.2440678] [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] [Revised: 10/09/2024] [Accepted: 12/04/2024] [Indexed: 12/20/2024] Open
Abstract
Liver diseases are a significant global health issue, characterized by elevated levels of disorder and death. The substantial impact of ageing on liver diseases and their prognosis is evident. Multiple processes are involved in the ageing process, which ultimately leads to functional deterioration of this organ. The process of liver ageing not only renders the liver more susceptible to diseases but also compromises the integrity of other organs due to the liver's critical function in metabolism regulation. A growing body of research suggests that long non-coding RNAs (lncRNAs) play a significant role in the majority of pathophysiological pathways. They regulate gene expression through a variety of interactions with microRNAs (miRNAs), messenger RNAs (mRNAs), DNA, or proteins. LncRNAs exert a major influence on the progression of age-related liver diseases through the regulation of cell proliferation, necrosis, apoptosis, senescence, and metabolic reprogramming. A concise overview of the current understanding of lncRNAs and their potential impact on the development of age-related liver diseases will be provided in this mini-review.
Collapse
Affiliation(s)
- Peyman Kheirandish Zarandi
- Department of Biology, Science and Research Branch, Islamic Azad University, Tehran, Iran
- Cancer Biology Signaling Pathway Interest Group (CBSPIG), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Mohsen Ghiasi
- Rajaie Cardiovascular Medical and Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Mohammad Heiat
- Baqiyatallah Research Center for Gastroenterology and Liver Diseases (BRCGL), Baqiyatallah University of Medical Sciences, Tehran, Iran
| |
Collapse
|
2
|
Fujii E, Ako J, Takahashi Y, Toda M, Iekushi K, Yamashita S. Serum Lipoprotein(a) Levels and Their Association with Atherosclerotic Cardiovascular Disease in Japan. J Atheroscler Thromb 2025; 32:421-438. [PMID: 39370270 PMCID: PMC11973527 DOI: 10.5551/jat.64953] [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: 05/24/2024] [Accepted: 08/19/2024] [Indexed: 10/08/2024] Open
Abstract
AIMS To investigate the distribution of lipoprotein(a) (Lp(a)) and its association with atherosclerotic cardiovascular disease (ASCVD) in Japanese patients at high risk for ASCVD using a health insurance database. METHODS Between July 2013 and June 2021, patients eligible for ASCVD prevention according to the 2017 Japan Atherosclerosis Society (JAS) guidelines with documented Lp(a) test results were extracted from the Medical Data Vision claims database and divided into three groups: primary prevention high-risk (Group I), secondary prevention (Group II) and secondary prevention high-risk (Group III). Data on lipid levels, cardiovascular morbidity risk factors and lipid-lowering treatments were extracted. RESULTS Of 700,580 patients with documented low-density lipoprotein cholesterol (LDL-C), 2,967 (0.42%) were tested for Lp(a). In 2,170 eligible patients, the median [interquartile range] serum concentration of Lp(a) was 13.9 [7.5-24.6] mg/dL, with 151 patients (7.0%) above the recommended risk threshold of ≥ 50 mg/dL. Lp(a) levels increased with risk across all prevention groups. Being in the highest Lp(a) quintile (Q5) was associated with an increased frequency of ASCVD (28.9% versus 18.9% in the lowest quintile (Q1) for unstable angina; 18.7% versus 10.1% for myocardial infarction; 27.9% versus 17.0% for ischemic stroke). In the secondary prevention groups, the proportion of patients meeting an LDL-C target of <70 mg/dL decreased from 30.2% in Q1 to 19.0% in Q5 for Group II and from 32.9% to 16.3% for Group III. CONCLUSIONS Despite a high prevalence of Lp(a) ≥ 50mg/dL in Japanese patients at high risk for ASCVD, it found that the Lp(a) testing rate was very low.
Collapse
Affiliation(s)
- Emi Fujii
- Medical Affairs Division, Novartis Pharma K.K
| | - Junya Ako
- Department of Cardiovascular Medicine, Kitasato University School of Medicine
| | | | | | | | | |
Collapse
|
3
|
Yang PT, Tang L, Guo HR, He YM, Qin YX, Yan L, Li ZX, Guo YZ, Wang JG. Prevalence of Lipoprotein(a) Measurement and its Association with Arteriosclerosis in Asymptomatic Individuals in China. J Atheroscler Thromb 2025; 32:513-524. [PMID: 39443133 PMCID: PMC11973526 DOI: 10.5551/jat.65214] [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: 06/20/2024] [Accepted: 09/08/2024] [Indexed: 10/25/2024] Open
Abstract
AIMS Lipoprotein(a) [Lp(a)] is an independent risk factor for atherosclerotic cardiovascular disease (ASCVD), and its level is genetically determined. Although guidelines and consensuses in various cardiovascular fields have emphasized the importance of Lp(a), screening for Lp(a) in China has not been well studied. METHODS A cross-sectional study was conducted using a random sample of 30,000 medical examiners from each of the five health check-up centres. The distribution of Lp(a) was described for those who completed Lp(a) testing, and logistic regression modelling was used to evaluate the relationship between Lp(a) levels and vascular structure and function in the population who underwent carotid ultrasound and brachial‒ankle pulse wave velocity (baPWV) measurements. RESULTS Lp(a) was measured in only 4400 (3.02%) of the 150,000 participants. Among those tested for Lp(a), the median concentration was 15.85 mg/dL. The proportion of participants with Lp(a) levels ≥ 30 mg/dL was 15.00%. Multiple logistic regression analysis revealed a significant correlation between Lp(a) and cIMT ≥ 1.0 mm (OR: 1.008, 95% CI: 1.001-1.014, P=0.020) and carotid artery plaques (OR: 1.010, 95% CI: 1.004-1.016, P=0.001) but no correlation with baPWV ≥ 1400 (OR: 0.999, 95% CI: 0.993-1.005, P=0.788) or baPWV ≥ 1800 (OR: 1.002, 95% CI: 0.993-1.011, P=0.634). CONCLUSIONS The detection rate of Lp(a) at health checkups is low, and Lp(a) is positively associated with cervical vascular sclerosis and plaque but not with baPWV. Therefore, the testing rate of Lp(a) and the awareness of the risk of vascular structural changes due to Lp(a) should be further improved.
Collapse
Affiliation(s)
- Ping-ting Yang
- Health Management Medical Center, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Li Tang
- Shaoxing Second Hospital, shaoxing, Zhejiang, China
| | - Hui-rong Guo
- Health Management Center, Changji Branch of The First Affiliated Hospital, Xinjiang Medical University, Changji, Xinjiang, China
| | - Yong-mei He
- Department of Health Management, Aerospace Center Hospital, Beijing, China
| | - Yue-xiang Qin
- Health Management Medical Center, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Lei Yan
- Department of General Practice CThe Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Zhen-xin Li
- Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Ya-zhang Guo
- Health Management Center, First Affiliated Hospital of Jishou University, JiShou, Hunan, China
| | - Jian-gang Wang
- Health Management Medical Center, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| |
Collapse
|
4
|
Nissen SE, Ni W, Shen X, Wang Q, Navar AM, Nicholls SJ, Wolski K, Michael L, Haupt A, Krege JH. Lepodisiran - A Long-Duration Small Interfering RNA Targeting Lipoprotein(a). N Engl J Med 2025. [PMID: 40162643 DOI: 10.1056/nejmoa2415818] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
BACKGROUND Elevated lipoprotein(a) concentrations are associated with atherosclerotic cardiovascular disease. The safety and efficacy of lepodisiran, an extended-duration, small interfering RNA targeting hepatic synthesis of lipoprotein(a), are unknown. METHODS We randomly assigned participants in a 1:2:2:2:2 ratio to receive lepodisiran at a dose of 16 mg, 96 mg, or 400 mg at baseline and again at day 180, lepodisiran at a dose of 400 mg at baseline and placebo at day 180, or placebo at baseline and at day 180, all administered by subcutaneous injection. Data from the two groups that received lepodisiran at a dose of 400 mg at baseline were pooled for the primary analysis. The primary end point was the time-averaged percent change from baseline in the serum lipoprotein(a) concentration (lepodisiran difference from placebo [i.e., placebo-adjusted]) during the period from day 60 to day 180. RESULTS A total of 320 participants underwent randomization; the median baseline lipoprotein(a) concentration was 253.9 nmol per liter. The placebo-adjusted time-averaged percent change from baseline in the serum lipoprotein(a) concentration from day 60 to day 180 was -40.8 percentage points (95% confidence interval [CI], -55.8 to -20.6) in the 16-mg lepodisiran group, -75.2 percentage points (95% CI, -80.4 to -68.5) in the 96-mg group, and -93.9 percentage points (95% CI, -95.1 to -92.5) in the pooled 400-mg groups. The corresponding change from day 30 to day 360 was -41.2 percentage points (95% CI, -55.4 to -22.4), -77.2 percentage points (95% CI, -81.8 to -71.5), -88.5 percentage points (95% CI, -90.8 to -85.6), and -94.8 percentage points (95% CI, -95.9 to -93.4) in the 16-mg, 96-mg, 400-mg-placebo, and 400-mg-400-mg dose groups, respectively. Serious adverse events, none of which were deemed by investigators to be related to lepodisiran or placebo, occurred in 35 participants. Dose-dependent, generally mild injection-site reactions occurred in up to 12% (8 of 69) of the participants in the highest lepodisiran dose group. CONCLUSIONS Lepodisiran reduced mean serum concentrations of lipoprotein(a) from 60 to 180 days after administration. (Funded by Eli Lilly; ALPACA ClinicalTrials.gov number, NCT05565742.).
Collapse
Affiliation(s)
- Steven E Nissen
- Cleveland Clinic Coordinating Center for Clinical Research, Cleveland
| | | | | | - Qiuqing Wang
- Cleveland Clinic Coordinating Center for Clinical Research, Cleveland
| | | | | | - Kathy Wolski
- Cleveland Clinic Coordinating Center for Clinical Research, Cleveland
| | | | | | | |
Collapse
|
5
|
Sheng L, Yang Y, Zhou Y. Association between lipoprotein(a) and diabetic peripheral neuropathy in patients with type 2 diabetes: a meta-analysis. Diabetol Metab Syndr 2025; 17:76. [PMID: 40033299 DOI: 10.1186/s13098-025-01621-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/24/2024] [Accepted: 01/31/2025] [Indexed: 03/05/2025] Open
Abstract
BACKGROUND Diabetic peripheral neuropathy (DPN) is a common complication of type 2 diabetes (T2D). Lipoprotein(a) [Lp(a)], a known cardiovascular risk factor, has been hypothesized to influence the development of DPN. This meta-analysis aimed to investigate the relationship between Lp(a) levels and DPN in patients with T2D. METHODS Following PRISMA 2020 guidelines, a systematic search of PubMed, Embase, Web of Science, Wanfang, and CNKI databases was performed up to October 12, 2024. Observational studies assessing blood Lp(a) levels in T2D patients with and without DPN or evaluating the association between Lp(a) and DPN risk were included. Data synthesis utilized a random-effects model to calculate standardized mean differences (SMDs) and odds ratios (ORs) with corresponding 95% confidence intervals (CIs). RESULTS Eleven studies with 18,022 patients were included. Patients with DPN had significantly higher Lp(a) levels than those without DPN (SMD: 0.10, 95% CI: 0.02-0.19, p = 0.01; I² = 43%). High Lp(a) levels were associated with DPN (OR: 1.31, 95% CI: 1.07-1.60, p = 0.009; I² = 62%). Subgroup analyses according to study design, mean age of the patients, methods for measuring Lp(a) concentration, cutoff values of a high Lp(a), and study quality scores showed consistent results (p for subgroup difference all > 0.05). A high Lp(a) was associated with DPN in studies from Asian countries, but not in those from European countries (p for subgroup difference = 0.001). CONCLUSION Elevated Lp(a) levels are associated DPN in T2D patients, particularly in studies from Asian countries.
Collapse
Affiliation(s)
- Li Sheng
- Department of Endocrinology, Affiliated Hospital of Nanjing University of Chinese Medicine, Changzhou Hospital of Traditional Chinese Medicine, No.25 Heping North Road, Changzhou, 213003, China
| | - Yiwen Yang
- Department of Endocrinology, Affiliated Hospital of Nanjing University of Chinese Medicine, Changzhou Hospital of Traditional Chinese Medicine, No.25 Heping North Road, Changzhou, 213003, China
| | - Yunqing Zhou
- Department of Endocrinology, Affiliated Hospital of Nanjing University of Chinese Medicine, Changzhou Hospital of Traditional Chinese Medicine, No.25 Heping North Road, Changzhou, 213003, China.
| |
Collapse
|
6
|
Clair V, Zirille FM, Gill E. Rethinking cardiovascular risk: The emerging role of lipoprotein(a) screening. Am J Prev Cardiol 2025; 21:100945. [PMID: 40103685 PMCID: PMC11919408 DOI: 10.1016/j.ajpc.2025.100945] [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: 11/17/2024] [Revised: 01/09/2025] [Accepted: 02/07/2025] [Indexed: 03/20/2025] Open
Abstract
Lipoprotein(a) [Lp(a)] is a genetically inherited, independent risk factor for cardiovascular disease (CVD), affecting approximately 20-25% of the global population. Elevated Lp(a) levels are associated with a 2-3-fold increased risk of myocardial infarction and aortic valve stenosis, comparable to the risk seen in individuals with familial hypercholesterolemia. Despite its clinical relevance, the integration of Lp(a) screening into routine practice has been limited by inconsistent measurement techniques and a lack of targeted treatments. Recent advancements, including improved assays and the development of potential Lp(a)-lowering therapies, have renewed focus on the importance of Lp(a) screening. This review aims to clarify the role of Lp(a) in cardiovascular health by examining current evidence on who should be screened, when screening should occur, and the most accurate methods for measuring Lp(a). Key recommendations include universal, one-time screening for adults, selective screening for high-risk pediatric patients, and special considerations for individuals with conditions such as familial hypercholesterolemia and chronic kidney disease. Advances in assay technology now allow for more precise Lp(a) measurement, supporting better risk stratification. Additionally, emerging therapies that specifically target elevated Lp(a) levels could lead to more personalized management of CVD risk. Our findings support the integration of Lp(a) screening into routine cardiovascular risk assessment, highlighting its potential to improve early detection and prevention strategies across diverse patient populations.
Collapse
Affiliation(s)
- Victoria Clair
- School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Francis M Zirille
- Department of Medicine, Division of Cardiology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Edward Gill
- Department of Medicine, Division of Cardiology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| |
Collapse
|
7
|
Velilla TA, Prendes CF, Grau NA, Calmarza P, Anguila SC, Estébanez BC, Castro MJC, Ceacero D, Martínez IG, Palencia MM, Foncillas JP, Román CR. 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
|
8
|
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. 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
|
9
|
Averna M, Cefalù AB. LP(a): The new marker of high cardiovascular risk. Nutr Metab Cardiovasc Dis 2025; 35:103845. [PMID: 39956697 DOI: 10.1016/j.numecd.2024.103845] [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: 10/24/2024] [Revised: 12/17/2024] [Accepted: 12/24/2024] [Indexed: 02/18/2025]
Abstract
The biological functions and role in human diseases of lipoprotein (a) discovered more than 60 years ago are still not fully understood. The high homology of apo(a) with plasminogen initially leads us to think of Lp(a) as a player in the coagulation system as pro-thrombotic factor. Over the years, a solid body of evidence from biology, epidemiology and from genetics and mendelian randomization has contributed to identify Lp(a) as a causal factor of atherosclerotic coronary heart disease, aortic calcific valve stenosis and ischaemic stroke. The active involvement of Lp(a) in atherogenesis and aortic calcific valve stenosis has been demonstrated by experimental data regarding the role of oxidized phospholipids, which are the cargo of Lp(a) and the presence of a Lp(a) receptor in valve interstitial cells. In secondary prevention, patients optimally treated for low density lipoprotein cholesterol (LDL-C) but with high Lp(a) levels show a residual cardiovascular risk. To date the LDL-C affecting drugs have a marginal effect on Lp(a). Statins produce a modest increase, monoclonal PCSK9i and Inclisiran a modest decrease not sufficient to reduce significantly the risk associated to Lp(a). Only lipoprotein apheresis and obicetrapib, a CETP novel inhibitor, reduce respectively by 75% and 40% Lp(a) levels. To obtain a lifetime risk reduction of 50% similar to that achieved by reducing LDL-C of about 40 mg/dl, Lp(a) should be reduced of about 100 mg/dl. The ongoing trials on drugs such as ASO, SiRnas, assembly inhibitors and maybe in the future the gene editing could obtain these results.
Collapse
Affiliation(s)
- Maurizio Averna
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties (PROMISE), Università degli Studi di Palermo, Via del Vespro 129, 90127, Palermo, Italy; Istituto di Biofisica, Consiglio Nazionale delle Ricerche, Via U. La Malfa 153, 90146, Palermo, Italy.
| | - Angelo B Cefalù
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties (PROMISE), Università degli Studi di Palermo, Via del Vespro 129, 90127, Palermo, Italy.
| |
Collapse
|
10
|
Antwi K, Downie P, Mbagaya W. Determination of the biological variation and reference change value of lipoprotein (a). Ann Clin Biochem 2025:45632251324063. [PMID: 39947649 DOI: 10.1177/00045632251324063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/27/2025]
Abstract
BACKGROUND Understanding lipoprotein (a) [Lp(a)] measurement variability is essential in establishing its coronary heart disease (CHD) association, and optimizing assessment and management of atherosclerotic cardiovascular disease (ASCVD) risk. We established the components of biological variation (BV) and reference change value (RCV) of Lp(a) in a UK cohort. METHOD 22 healthy individuals were recruited to the study. Blood samples were collected for six consecutive weeks and analysed in duplicate using the Lp(a) assay by Sentinel Diagnostics on the Beckman Coulter AU5800. Outlier, heterogeneity, normality, and trend analysis were performed, followed by CV-ANOVA to determine estimates of BV, adhering to the 14 BIVAC quality items. RCV was calculated based on estimated CVA and CVI. RESULTS Four participants were excluded from the analysis as their mean Lp(a) levels fell below the functional sensitivity of the assay. Mean Lp(a) concentration ranged from 14 to 241 nmol/L. The overall estimate of CVI for all participants was 10.9% (95% CI of 9.1 - 13.0%). The RCV for Lp(a) was +31.6%/-24.0%. CONCLUSION Our study obtained a CVI estimate for Lp(a) that aligned consistently with recent studies adhering to the quality specifications outlined in the BIVAC checklist. The CVI estimate was significantly lower than Lp(a) estimates reported in studies up to 2003. The CVI estimate highlights the limitations of relying solely on a single Lp(a) measurement for prognosticating ASCVD risk and identifying candidates for novel Lp(a) therapies, particularly when the measured value is near clinical decision thresholds.
Collapse
Affiliation(s)
- Kofi Antwi
- Department of Clinical Biochemistry, University Hospitals Bristol and Weston NHS Foundation Trust, Bristol, UK
| | - Paul Downie
- Department of Clinical Biochemistry, University Hospitals Bristol and Weston NHS Foundation Trust, Bristol, UK
| | - Wycliffe Mbagaya
- Department of Clinical Biochemistry, University Hospitals Bristol and Weston NHS Foundation Trust, Bristol, UK
| |
Collapse
|
11
|
Greco A, Finocchiaro S, Spagnolo M, Faro DC, Mauro MS, Raffo C, Sangiorgio G, Imbesi A, Laudani C, Mazzone PM, Ammirabile N, Giacoppo D, Landolina D, Capodanno D. Lipoprotein(a) as a Pharmacological Target: Premises, Promises, and Prospects. Circulation 2025; 151:400-415. [PMID: 39928714 DOI: 10.1161/circulationaha.124.069210] [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: 02/11/2024] [Accepted: 10/29/2024] [Indexed: 02/12/2025]
Abstract
Atherosclerotic cardiovascular disease is a major health concern worldwide and requires effective preventive measures. Lp(a) (lipoprotein [a]) has recently garnered attention as an independent risk factor for astherosclerotic cardiovascular disease, with proinflammatory and prothrombotic mechanisms contributing to its atherogenicity. On an equimolar basis, Lp(a) is ~5 to 6 times more atherogenic than particles that have been widely associated with adverse cardiovascular outcomes, such as LDL (low-density lipoprotein). Lp(a) can enter the vessel wall, leading to the accumulation of oxidized phospholipids in the arterial intima, which are crucial for initiating plaque inflammation and triggering vascular disease progression. In addition, Lp(a) may cause atherothrombosis through interactions between apoA (apolipoprotein A) and the platelet PAR-1 (protease-activated receptor 1) receptor, as well as competitive inhibition of plasminogen. Because Lp(a) is mostly determined on genetic bases, a 1-time assessment in a lifetime can suffice to identify patients with elevated levels. Mendelian randomization studies and post hoc analyses of randomized trials of LDL cholesterol-lowering drugs showed a causal link between Lp(a) concentrations and cardiovascular outcomes, with therapeutic reduction of Lp(a) expected to contribute to estimated cardiovascular risk mitigation. Many Lp(a)-lowering drugs, including monoclonal antibodies, small interfering ribonucleic acids, antisense oligonucleotides, small molecules, and gene editing compounds, are at different stages of clinical investigation and show promise for clinical use. In particular, increased Lp(a) testing and treatment are expected to have a substantial impact at the population level, enabling the identification of high-risk individuals and the subsequent prevention of a large number of cardiovascular events. Ongoing phase 3 trials will further elucidate the cardiovascular benefits of Lp(a) reduction over the long term, offering potential avenues for targeted interventions and improved cardiovascular outcomes.
Collapse
Affiliation(s)
- Antonio Greco
- Division of Cardiology, Azienda Ospedaliero-Universitaria Policlinico "G. Rodolico-San Marco," University of Catania, Italy
| | - Simone Finocchiaro
- Division of Cardiology, Azienda Ospedaliero-Universitaria Policlinico "G. Rodolico-San Marco," University of Catania, Italy
| | - Marco Spagnolo
- Division of Cardiology, Azienda Ospedaliero-Universitaria Policlinico "G. Rodolico-San Marco," University of Catania, Italy
| | - Denise Cristiana Faro
- Division of Cardiology, Azienda Ospedaliero-Universitaria Policlinico "G. Rodolico-San Marco," University of Catania, Italy
| | - Maria Sara Mauro
- Division of Cardiology, Azienda Ospedaliero-Universitaria Policlinico "G. Rodolico-San Marco," University of Catania, Italy
| | - Carmelo Raffo
- Division of Cardiology, Azienda Ospedaliero-Universitaria Policlinico "G. Rodolico-San Marco," University of Catania, Italy
| | - Giuseppe Sangiorgio
- Division of Cardiology, Azienda Ospedaliero-Universitaria Policlinico "G. Rodolico-San Marco," University of Catania, Italy
| | - Antonino Imbesi
- Division of Cardiology, Azienda Ospedaliero-Universitaria Policlinico "G. Rodolico-San Marco," University of Catania, Italy
| | - Claudio Laudani
- Division of Cardiology, Azienda Ospedaliero-Universitaria Policlinico "G. Rodolico-San Marco," University of Catania, Italy
| | - Placido Maria Mazzone
- Division of Cardiology, Azienda Ospedaliero-Universitaria Policlinico "G. Rodolico-San Marco," University of Catania, Italy
| | - Nicola Ammirabile
- Division of Cardiology, Azienda Ospedaliero-Universitaria Policlinico "G. Rodolico-San Marco," University of Catania, Italy
| | - Daniele Giacoppo
- Division of Cardiology, Azienda Ospedaliero-Universitaria Policlinico "G. Rodolico-San Marco," University of Catania, Italy
| | - Davide Landolina
- Division of Cardiology, Azienda Ospedaliero-Universitaria Policlinico "G. Rodolico-San Marco," University of Catania, Italy
| | - Davide Capodanno
- Division of Cardiology, Azienda Ospedaliero-Universitaria Policlinico "G. Rodolico-San Marco," University of Catania, Italy
| |
Collapse
|
12
|
Moyá Amengual A, Serrano-Cumplido A. [Lp(a): What we know, what we don't know and what we hope for]. Semergen 2025; 51:102451. [PMID: 39922183 DOI: 10.1016/j.semerg.2025.102451] [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: 12/03/2024] [Revised: 01/09/2025] [Accepted: 01/12/2025] [Indexed: 02/10/2025]
Abstract
There is no doubt that lipoprotein(a) [Lp(a)] is a structurally complex molecule with unique biological functions. It plays an important role in the inflammatory process through multiple mechanisms, contributes to endothelial dysfunction, activation of monocytes, macrophages and proliferation of smooth muscle cells, and promotes the development of atherosclerotic cardiovascular disease (ASCVD). It is important to point out the complex bidirectional relationship between Lp(a) and inflammation, influencing one another and even exerting anti-inflammatory effects in certain situations. Likewise, Lp(a) can favor the development of heart valve disease, especially of the aortic valve. Numerous publications emphasize the need to determine Lp(a) levels in the population at least once in life and possible strategies to mitigate the risk of ASCVD generated by high Lp(a) levels. However, doubts or lack of knowledge persist about the need to measure this parameter, either due to the uncertainty of how to manage patients with high levels of Lp(a), due to insufficient knowledge about its physiological function or because its levels persist unchanged, to a large extent, throughout life as the genetic character of this molecule takes precedence. On the other hand, there are still no specific approved therapies that reduce its levels and arouse sufficient interest for its management. However, many societies, such as the European Society of Cardiology (SEC) or the Spanish Society of Atherosclerosis (SEA), raise the need to determine Lp(a) and intensive management of cardiovascular risk factors in patients with high Lp(a) levels along with therapies that mitigate the associated ASCVD risk. Likewise, the identification of high levels of Lp(a) offers the opportunity to screen family members, better control of cardiovascular risk and the possibility of developing clinical trials that profile individual and population risk that allow for more personalized actions.
Collapse
Affiliation(s)
- A Moyá Amengual
- Servei de Salut de les Illes Balears, Palma de Mallorca, España
| | | |
Collapse
|
13
|
Bhatia HS, Wandel S, Willeit P, Lesogor A, Bailey K, Ridker PM, Nestel P, Simes J, Tonkin A, Schwartz GG, Colhoun H, Wanner C, Tsimikas S. Independence of Lipoprotein(a) and Low-Density Lipoprotein Cholesterol-Mediated Cardiovascular Risk: A Participant-Level Meta-Analysis. Circulation 2025; 151:312-321. [PMID: 39492722 PMCID: PMC11771346 DOI: 10.1161/circulationaha.124.069556] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Accepted: 09/24/2024] [Indexed: 11/05/2024]
Abstract
BACKGROUND Low-density lipoprotein cholesterol (LDL-C) and lipoprotein(a) (Lp[a]) levels are independently associated with atherosclerotic cardiovascular disease (ASCVD). However, the relationship between Lp(a) level, LDL-C level, and ASCVD risk at different thresholds is not well defined. METHODS A participant-level meta-analysis of 27 658 participants enrolled in 6 placebo-controlled statin trials was performed to assess the association of LDL-C and Lp(a) levels with risk of fatal or nonfatal coronary heart disease events, stroke, or any coronary or carotid revascularization (ASCVD). The multivariable-adjusted association between baseline Lp(a) level and ASCVD risk was modeled continuously using generalized additive models, and the association between baseline LDL-C level and ASCVD risk by baseline Lp(a) level by Cox proportional hazards models with random effects. The joint association between Lp(a) level and statin-achieved LDL-C level with ASCVD risk was evaluated using Cox proportional hazards models. RESULTS Compared with an Lp(a) level of 5 mg/dL, increasing levels of Lp(a) were log-linearly associated with ASCVD risk in statin- and placebo-treated patients. Among statin-treated individuals, those with Lp(a) level >50 mg/dL (≈125 nmol/L) had increased risk across all quartiles of achieved LDL-C level and absolute change in LDL-C level. Even among those with the lowest quartile of achieved LDL-C level (3.1-77.0 mg/dL), those with Lp(a) level >50 mg/dL had greater ASCVD risk (hazard ratio, 1.38 [95% CI, 1.06-1.79]) than those with Lp(a) level ≤50 mg/dL. The greatest risk was observed with both Lp(a) level >50 mg/dL and LDL-C level in the fourth quartile (hazard ratio, 1.90 [95% CI, 1.46-2.48]). CONCLUSIONS These findings demonstrate the independent and additive nature of Lp(a) and LDL-C levels for ASCVD risk, and that LDL-C lowering does not fully offset Lp(a)-mediated risk.
Collapse
Affiliation(s)
- Harpreet S. Bhatia
- Division of Cardiology, Department of Medicine, University of California, San Diego, La Jolla (H.S.B., S.T.)
| | - Simon Wandel
- Novartis Pharma AG, Basel, Switzerland (S.W., A.L., K.B.)
| | - Peter Willeit
- Department of Medical Statistics, Informatics and Health Economics at the Medical University of Innsbruck, Austria (P.W.)
| | | | - Keith Bailey
- Novartis Pharma AG, Basel, Switzerland (S.W., A.L., K.B.)
| | - Paul M. Ridker
- Brigham and Women’s Hospital, Harvard Medical School, Boston, MA (P.M.R.)
| | - Paul Nestel
- Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia (P.N.)
| | - John Simes
- NHMRC Clinical Trials Centre, University of Sydney, NSW, Australia (J.S.)
| | - Andrew Tonkin
- School of Public Health and Preventive Medicine, Monash University, Melbourne, Victoria, Australia (A.T.)
| | - Gregory G. Schwartz
- Division of Cardiology, Rocky Mountain Regional VA Medical Center and University of Colorado School of Medicine, Aurora (G.G.S.)
| | - Helen Colhoun
- MRC Human Genetics Unit, Centre for Genomic and Experimental Medicine, MRC Institute of Genetics & Molecular Medicine, Edinburgh, UK (H.C.)
| | - Christoph Wanner
- Division of Nephrology, Department of Internal Medicine and Comprehensive Heart Failure Centre, University Hospital of Würzburg, Germany (C.W.)
| | - Sotirios Tsimikas
- Division of Cardiology, Department of Medicine, University of California, San Diego, La Jolla (H.S.B., S.T.)
| |
Collapse
|
14
|
Konieczyńska M, Natorska J, Ząbczyk M, Undas A. Lipoprotein(a) and thromboembolism: current state of knowledge and unsolved issues. Arch Med Sci 2024; 20:1770-1783. [PMID: 39967936 PMCID: PMC11831339 DOI: 10.5114/aoms/197357] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/06/2024] [Accepted: 12/13/2024] [Indexed: 02/20/2025] Open
Abstract
Lipoprotein(a) [Lp(a)], a low-density lipoprotein-like particle containing a highly polymorphic apolipoprotein(a) [apo(a)] homologous in > 80% to plasminogen, was identified as a genetically determined independent risk factor for cardiovascular disease. Elevated Lp(a) levels, found in about 20% of Europeans, are strongly linked to higher rates of myocardial infarction, major adverse cardiac events, accelerated plaque progression, ischemic stroke (especially in younger adults), and calcific aortic valve disease. However, its role in venous thromboembolism, including atypical locations like cerebral and retinal vein thrombosis, remains controversial despite several shared mechanisms underlying arterial and venous thromboembolism. The most robust evidence supports antifibrinolytic properties of elevated Lp(a), particularly smaller apo(a) isoforms, which inhibit plasminogen activation mainly by interacting with the tissue-type plasminogen activator, plasminogen, and fibrin. Other prothrombotic mechanisms include increased synthesis of plasminogen activator inhibitor (PAI-1), formation of denser fibrin networks composed of thinner fibers, less susceptible to lysis, increased platelet activation, enhanced oxidation of phospholipids leading to a low-grade proinflammatory state, upregulated tissue factor expression, and suppression of tissue factor pathway inhibitor. Targeted Lp(a) lowering therapies are currently being tested in randomized clinical trials and could potentially have clinically relevant antithrombotic effects, evidenced by the reduced risk of thromboembolism. This review summarizes the available data on the prothrombotic and antifibrinolytic actions of Lp(a), along with clinical evidence for the increased risk of thromboembolic events related to elevated Lp(a). It also introduces new concepts to explain discrepant clinical results regarding venous events, highlighting the impact of oxidized phospholipids on a prothrombotic state under conditions of high Lp(a).
Collapse
Affiliation(s)
- Małgorzata Konieczyńska
- Department of Thromboembolic Disorders, Institute of Cardiology, Jagiellonian University Medical College, Krakow, Poland
- The John Paul II Hospital, Krakow, Poland
| | - Joanna Natorska
- Department of Thromboembolic Disorders, Institute of Cardiology, Jagiellonian University Medical College, Krakow, Poland
- The John Paul II Hospital, Krakow, Poland
| | - Michał Ząbczyk
- Department of Thromboembolic Disorders, Institute of Cardiology, Jagiellonian University Medical College, Krakow, Poland
- The John Paul II Hospital, Krakow, Poland
| | - Anetta Undas
- Department of Thromboembolic Disorders, Institute of Cardiology, Jagiellonian University Medical College, Krakow, Poland
- The John Paul II Hospital, Krakow, Poland
| |
Collapse
|
15
|
Letsiou S, Damigou E, Nomikos T, Pergantis SA, Pitsavos C, Panagiotakos D, Antonopoulou S. Deciphering the associations of selenium distribution in serum GPx-3 and selenoprotein P with cardiovascular risk factors in a healthy population with moderate levels of selenium: The ATTICA study. J Trace Elem Med Biol 2024; 86:127509. [PMID: 39190954 DOI: 10.1016/j.jtemb.2024.127509] [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: 06/18/2024] [Revised: 08/14/2024] [Accepted: 08/14/2024] [Indexed: 08/29/2024]
Abstract
BACKGROUND Selenium (Se) is an essential micronutrient, important for human health. The relationship of Se with cardiovascular risk factors is still inconclusive, especially regarding the role of different selenoproteins. The present study evaluated the relation of total serum Se as well as its distribution in plasma selenoproteins, namely glutathione peroxidase 3 (GPx3) and selenoprotein P (SelP) with cardiovascular risk factors in a sex-specific manner, in a healthy population with moderate levels of Se. METHODS A sub-sample from the ATTICA Study's database, consisting of 398 participants (160 females and 238 males) with data on Se and selenoproteins levels, was considered. GPx3, SelP and the main non-specific serum selenium containing protein, selenoalbumin (SeAlb) were simultaneously determined in human plasma by high-performance liquid chromatography (HPLC) coupled with inductively coupled plasma mass spectrometry (ICP-MS) at baseline. RESULTS Participants that belong to the highest tertiles of GPx3 and SelP presented the lowest blood pressure. Homocysteine was inversely associated with SelP and its ratio SelP/TSe in both sexes. In males, the lowest tertile of GPx3 showed lower adiponectin levels (0.66 ± 0.21 μg/mL) in comparison to the 2nd tertile of GPx3 (p=0.002), SelP was inversely associated with visceral adipose index (VAI) (-2.29 ± 0.81, p=0.005). Particularly, in males, the middle tertile of SelP had the lowest VAI values. Regarding females, lower Lp(a) concentration by 11.96 ± 5.84 mg/dL was observed in low SelP levels while higher leptin concentration by 2.30 ± 0.73 μg/L and lower fibrinogen concentration by 27.32 ± 13.30 mg/dL was detected in low GPx3 levels. CONCLUSION Circulating selenoproteins exert differentiated effects on cardiovascular risk factors, some of them in a sex-specific manner.
Collapse
Affiliation(s)
- Sophia Letsiou
- Department of Nutrition and Dietetics, School of Health Sciences and Education, Harokopio University, Athens, Greece
| | - Evangelia Damigou
- Department of Nutrition and Dietetics, School of Health Sciences and Education, Harokopio University, Athens, Greece
| | - Tzortzis Nomikos
- Department of Nutrition and Dietetics, School of Health Sciences and Education, Harokopio University, Athens, Greece
| | | | - Christos Pitsavos
- First Cardiology Clinic, Medical School, National and Kapodistrian University of Athens, Hippokration Hospital, Athens, Greece
| | - Demosthenes Panagiotakos
- Department of Nutrition and Dietetics, School of Health Sciences and Education, Harokopio University, Athens, Greece
| | - Smaragdi Antonopoulou
- Department of Nutrition and Dietetics, School of Health Sciences and Education, Harokopio University, Athens, Greece.
| |
Collapse
|
16
|
Gruber I, Kollerits B, Forer L, Di Maio S, Schachtl-Riess JF, Kheirkhah A, Schönherr S, Schultheiss UT, Köttgen A, Eckardt KU, Coassin S, Lamina C, Kronenberg F. Lipoprotein(a) concentrations and cardiovascular disease in patients with chronic kidney disease: Results from the German Chronic Kidney Disease study. J Intern Med 2024; 296:510-526. [PMID: 39513193 DOI: 10.1111/joim.20027] [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] [Indexed: 11/15/2024]
Abstract
BACKGROUND Lipoprotein(a) (Lp(a)) is a causal, genetically determined risk factor for cardiovascular disease (CVD) in the general population. Patients with chronic kidney disease (CKD) have an increased CVD risk and elevated Lp(a) concentrations. Only a few studies on Lp(a) were performed in persons with mild-to-moderate CKD; none of them used genetic variants to explore potential causal associations. OBJECTIVES This study aims to investigate the association of measured and genetically predicted Lp(a) concentrations on prevalent and incident CVD events in the German Chronic Kidney Disease (GCKD) study. METHODS The study included 5043 participants of European ancestry with an estimated glomerular filtration rate (eGFR) between 30 and 60 mL/min/1.73 m2 or an eGFR >60 mL/min/1.73 m2 in the presence of overt albuminuria with a follow-up of 6.5 years. RESULTS With each 10 mg/dL higher Lp(a) concentration, odds for prevalent CVD (1290 events) increased 1.065-fold (95%CI: 1.042-1.088, p < 0.001). The risk was significantly higher in patients with Lp(a) ≥50 mg/dL but most pronounced in Lp(a) ≥70 mg/dL (odds ratio = 1.775 [1.409-2.231], p < 0.001) compared to Lp(a) <30 mg/dL. Each 10 mg/dL higher Lp(a) concentration and Lp(a) ≥70 mg/dL increased the risk for incident 3-point major adverse cardiovascular events (MACEs) (474 events): hazard ratio [HR] = 1.037 [1.009-1.067], p = 0.009 and HR = 1.335 [1.001-1.781], p = 0.050), respectively. Similar results were obtained for 4-point MACE (653 events). Analyses based on apo(a) isoforms and genetically predicted Lp(a) concentrations led to even stronger associations. CONCLUSIONS In patients with mild-to-severe CKD, elevated Lp(a) concentrations and genetic determinants of Lp(a) concentrations are significantly associated with CVD at baseline and during follow-up, independent of traditional risk factors.
Collapse
Affiliation(s)
- Ida Gruber
- Institute of Genetic Epidemiology, Medical University of Innsbruck, Innsbruck, Austria
| | - Barbara Kollerits
- Institute of Genetic Epidemiology, Medical University of Innsbruck, Innsbruck, Austria
| | - Lukas Forer
- Institute of Genetic Epidemiology, Medical University of Innsbruck, Innsbruck, Austria
| | - Silvia Di Maio
- Institute of Genetic Epidemiology, Medical University of Innsbruck, Innsbruck, Austria
| | | | - Azin Kheirkhah
- Institute of Genetic Epidemiology, Medical University of Innsbruck, Innsbruck, Austria
| | - Sebastian Schönherr
- Institute of Genetic Epidemiology, Medical University of Innsbruck, Innsbruck, Austria
| | - Ulla T Schultheiss
- Faculty of Medicine and Medical Center, Institute of Genetic Epidemiology, University of Freiburg, Freiburg, Germany
- Department of Medicine IV, Nephrology and Primary Care, Faculty of Medicine and Medical Center, University of Freiburg, Freiburg, Germany
- Synlab MVZ Humangenetik Freiburg GmbH, Freiburg, Germany
| | - Anna Köttgen
- Faculty of Medicine and Medical Center, Institute of Genetic Epidemiology, University of Freiburg, Freiburg, Germany
| | - Kai-Uwe Eckardt
- Department of Nephrology and Hypertension, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
- Department of Nephrology and Medical Intensive Care, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Stefan Coassin
- Institute of Genetic Epidemiology, Medical University of Innsbruck, Innsbruck, Austria
| | - Claudia Lamina
- Institute of Genetic Epidemiology, Medical University of Innsbruck, Innsbruck, Austria
| | - Florian Kronenberg
- Institute of Genetic Epidemiology, Medical University of Innsbruck, Innsbruck, Austria
| |
Collapse
|
17
|
Giannakopoulou SP, Chrysohoou C, Antonopoulou S, Damigou E, Barkas F, Vafia C, Kravvariti E, Tsioufis C, Pitsavos C, Liberopoulos E, Sfikakis PP, Panagiotakos D. Discrimination and net-reclassification of cardiovascular disease risk with lipoprotein(a) levels: The ATTICA study (2002-2022). J Clin Lipidol 2024; 18:e957-e967. [PMID: 38908971 DOI: 10.1016/j.jacl.2024.04.126] [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: 03/19/2024] [Revised: 04/12/2024] [Accepted: 04/15/2024] [Indexed: 06/24/2024]
Abstract
BACKGROUND Lipoprotein(a) [Lp(a)] is recognized as a risk factor for atherosclerotic cardiovascular disease (ASCVD). However, its influence on clinical risk evaluations remains unclear. OBJECTIVE This study aimed to determine whether Lp(a) improves CVD risk prediction among apparently healthy adults from the general population. METHODS In 2002, n = 3,042 adults free of CVD, residing in the Athens metropolitan area, in Greece, were recruited. A 20-year follow-up was conducted in 2022, comprising n = 2,169 participants, of which n = 1,988 had complete data for CVD incidence. RESULTS Lp(a) levels were significantly associated with 20-year ASCVD incidence in the crude model (hazard ratio per 1 mg/dL: 1.004, p = 0.048), but not in multi-adjusted models considering demographic, lifestyle, and clinical factors. Adding Lp(a) to the Reynolds Risk Score (RRS) and Framingham Risk Score (FRS) variables resulted in positive net reclassification improvement (NRI) values (0.159 and 0.160 respectively), indicating improved risk classification. Mediation analysis suggested that C-reactive protein, interleukin-6, and fibrinogen mediate the relationship between Lp(a) and ASCVD. No significant interaction was observed between Lp(a) and potential moderators. CONCLUSION Lp(a) levels can predict 20-year CVD outcomes and improve CVD risk prediction within the general population, possibly via the intricate relationship between Lp(a), systemic inflammation, atherothrombosis.
Collapse
Affiliation(s)
- Sofia-Panagiota Giannakopoulou
- Department of Nutrition and Dietetics (Drs Giannakopoulou, Antonopoulou, Damigou, Vafia and Panagiotakos), School of Health Sciences and Education, Harokopio University, 17676 Athens, Greece
| | - Christina Chrysohoou
- First Cardiology Clinic (Drs Chrysohoou, Tsioufis and Pitsavos), Medical School, National and Kapodistrian University of Athens, Hippokration Hospital, 15772 Athens, Greece
| | - Smaragdi Antonopoulou
- Department of Nutrition and Dietetics (Drs Giannakopoulou, Antonopoulou, Damigou, Vafia and Panagiotakos), School of Health Sciences and Education, Harokopio University, 17676 Athens, Greece
| | - Evangelia Damigou
- Department of Nutrition and Dietetics (Drs Giannakopoulou, Antonopoulou, Damigou, Vafia and Panagiotakos), School of Health Sciences and Education, Harokopio University, 17676 Athens, Greece
| | - Fotios Barkas
- Department of Internal Medicine (Dr Barkas), Medical School, University of Ioannina, 45500 Ioannina, Greece
| | - Christina Vafia
- Department of Nutrition and Dietetics (Drs Giannakopoulou, Antonopoulou, Damigou, Vafia and Panagiotakos), School of Health Sciences and Education, Harokopio University, 17676 Athens, Greece
| | - Evrydiki Kravvariti
- First Department of Propaedeutic Internal Medicine (Drs Kravvariti, Liberopoulos and Sfikakis), Medical School, National and Kapodistrian University of Athens, Laiko General Hospital, 15772 Athens, Greece
| | - Costas Tsioufis
- First Cardiology Clinic (Drs Chrysohoou, Tsioufis and Pitsavos), Medical School, National and Kapodistrian University of Athens, Hippokration Hospital, 15772 Athens, Greece
| | - Christos Pitsavos
- First Cardiology Clinic (Drs Chrysohoou, Tsioufis and Pitsavos), Medical School, National and Kapodistrian University of Athens, Hippokration Hospital, 15772 Athens, Greece
| | - Evangelos Liberopoulos
- First Department of Propaedeutic Internal Medicine (Drs Kravvariti, Liberopoulos and Sfikakis), Medical School, National and Kapodistrian University of Athens, Laiko General Hospital, 15772 Athens, Greece
| | - Petros P Sfikakis
- First Department of Propaedeutic Internal Medicine (Drs Kravvariti, Liberopoulos and Sfikakis), Medical School, National and Kapodistrian University of Athens, Laiko General Hospital, 15772 Athens, Greece
| | - Demosthenes Panagiotakos
- Department of Nutrition and Dietetics (Drs Giannakopoulou, Antonopoulou, Damigou, Vafia and Panagiotakos), School of Health Sciences and Education, Harokopio University, 17676 Athens, Greece.
| |
Collapse
|
18
|
Schuth U, Gill K, Telyuk P, Bawamia BR, Austin D, Zaman A. Role of Lipoprotein(a) Reduction in Cardiovascular Disease. J Clin Med 2024; 13:6311. [PMID: 39518451 PMCID: PMC11546410 DOI: 10.3390/jcm13216311] [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: 09/30/2024] [Accepted: 10/14/2024] [Indexed: 11/16/2024] Open
Abstract
Recent studies have shown that lipoprotein(a) (Lp(a)) is an important risk factor for a plethora of different cardiovascular diseases. It has been proven that Lp(a) levels are genetically determined and correlate with risk of cardiovascular disease, independent of lifestyle factors. As of yet, treatment options to reduce Lp(a) levels are limited, but new research into Lp(a) reduction yields promising results. This review delves into Lp(a)'s biochemistry and mechanism of effect, the association between Lp(a) and cardiovascular diseases, and possible therapies to minimise cardiovascular disease.
Collapse
Affiliation(s)
- Uma Schuth
- Faculty of Medicine, St Andrews University, St Andrews KY16 9ST, UK;
| | - Kieran Gill
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne NE2 4AX, UK;
| | - Pyotr Telyuk
- Academic Cardiovascular Unit, The James Cook University Hospital, South Tees Hospitals NHS Foundation Trust, Middlesbrough TS4 3BW, UK; (P.T.); (D.A.)
| | | | - David Austin
- Academic Cardiovascular Unit, The James Cook University Hospital, South Tees Hospitals NHS Foundation Trust, Middlesbrough TS4 3BW, UK; (P.T.); (D.A.)
- Population Health Sciences Institute, Newcastle University, Newcastle-upon-Tyne NE2 4AX, UK
| | - Azfar Zaman
- Cardiology, Freeman Hospital and Vascular Biology and Medicine, Newcastle University, Newcastle upon Tyne NE2 4AX, UK
| |
Collapse
|
19
|
Boffa MB, Koschinsky ML. Lipoprotein(a) and cardiovascular disease. Biochem J 2024; 481:1277-1296. [PMID: 39302109 PMCID: PMC11555715 DOI: 10.1042/bcj20240037] [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: 07/03/2024] [Revised: 08/28/2024] [Accepted: 08/28/2024] [Indexed: 09/22/2024]
Abstract
Elevated plasma levels of lipoprotein(a) (Lp(a)) are a prevalent, independent, and causal risk factor for atherosclerotic cardiovascular disease and calcific aortic valve disease. Lp(a) consists of a lipoprotein particle resembling low density lipoprotein and the covalently-attached glycoprotein apolipoprotein(a) (apo(a)). Novel therapeutics that specifically and potently lower Lp(a) levels are currently in advanced stages of clinical development, including in large, phase 3 cardiovascular outcomes trials. However, fundamental unanswered questions remain concerning some key aspects of Lp(a) biosynthesis and catabolism as well as the true pathogenic mechanisms of the particle. In this review, we describe the salient biochemical features of Lp(a) and apo(a) and how they underlie the disease-causing potential of Lp(a), the factors that determine plasma Lp(a) concentrations, and the mechanism of action of Lp(a)-lowering drugs.
Collapse
Affiliation(s)
- Michael B. Boffa
- Department of Biochemistry, University of Western Ontario, London, Ontario, Canada
- Robarts Research Institute, University of Western Ontario, London, Ontario, Canada
| | - Marlys L. Koschinsky
- Robarts Research Institute, University of Western Ontario, London, Ontario, Canada
- Department of Physiology and Pharmacology, University of Western Ontario, London, Ontario, Canada
| |
Collapse
|
20
|
Dalla Vestra M, Grolla E, Autiero G, Presotto F. Peripheral artery disease risk factors: A focus on lipoprotein(a). Arch Cardiovasc Dis 2024; 117:584-589. [PMID: 39227282 DOI: 10.1016/j.acvd.2024.07.061] [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: 02/20/2024] [Revised: 07/04/2024] [Accepted: 07/14/2024] [Indexed: 09/05/2024]
Abstract
There is a well-established and strong link between high lipoprotein(a) concentration and coronary heart disease, but the evidence regarding peripheral artery disease and carotid atherosclerosis is not as conclusive. This review aims to summarize the relationships between lipoprotein(a), peripheral artery disease and carotid atherosclerosis, in order to try to understand the weight of lipoprotein(a) in determining the development, progression and any complications of atherosclerotic plaque at the carotid and peripheral artery level. There is currently no effective therapy to reduce lipoprotein(a) concentration, but understanding its significance as a vascular risk factor is the starting point to then explore (when effective therapies become available) if there is the possibility, even in patients with peripheral artery disease and carotid atherosclerosis, to achieve better control of the residual vascular risk that is ultimately induced by lipoprotein(a).
Collapse
Affiliation(s)
- Michele Dalla Vestra
- Department of Internal Medicine, Ospedale dell'Angelo, 30174 Mestre-Venezia, Italy.
| | - Elisabetta Grolla
- Department of Cardiology, Ospedale dell'Angelo, 30174 Mestre-Venezia, Italy
| | - Giuliana Autiero
- Department of Internal Medicine, Ospedale dell'Angelo, 30174 Mestre-Venezia, Italy
| | - Fabio Presotto
- Department of Internal Medicine, Ospedale dell'Angelo, 30174 Mestre-Venezia, Italy
| |
Collapse
|
21
|
Buciu IC, Tieranu EN, Pircalabu AS, Zlatian OM, Donoiu I, Militaru C, Militaru S, Militaru C. The Relationship between Lipoprotein A and the Prevalence of Multivessel Coronary Artery Disease in Young Patients with Acute Myocardial Infarction: An Observational Study. Biomedicines 2024; 12:2159. [PMID: 39335672 PMCID: PMC11429128 DOI: 10.3390/biomedicines12092159] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2024] [Revised: 09/16/2024] [Accepted: 09/20/2024] [Indexed: 09/30/2024] Open
Abstract
Introduction: Cardiovascular diseases are the leading cause of mortality worldwide, with a significant impact on socioeconomic aspects. Various biomarkers have been studied in relation to the diagnosis, progression, and prognosis of atherosclerotic disease, with lipoprotein (a) [Lp (a)] standing out as an important predictor of cardiovascular risk. This observational study aimed to clarify the association between Lp (a) levels and the severity of significant multivessel coronary lesions in acute myocardial infarction (AMI) patients. Materials and Methods: Conducted at the Clinical Emergency County Hospital of Craiova, Romania, the study involved 256 young patients divided into two groups based on Lp (a) levels: Group A (Lp (a) < 30 mg/dL) and Group B (Lp (a) ≥ 30 mg/dL). Patients included young adults up to 55 years for males and 60 years for females, excluding those with familial hypercholesterolemia. Results: The study revealed a significant association between elevated Lp (a) levels and the presence of multivessel coronary lesions. Patients with Lp (a) concentrations ≥ 30 mg/dL exhibited a higher prevalence of multivessel disease compared to those with lower levels. Discussion: The findings suggest that elevated Lp (a) levels are a crucial biomarker for the risk of coronary artery disease, particularly in young patients with AMI. The study emphasizes the need for aggressive lipid management strategies and personalized treatment approaches, considering the significant role of Lp (a) in atherosclerosis and AMI. Conclusions: Lipoprotein A levels above 30 mg/dL are associated with a higher prevalence of multivessel coronary lesions. Multivariate analysis revealed that higher Lp (a) levels and lower HDL levels are linked to an increased risk of multivessel coronary lesions.
Collapse
Affiliation(s)
- Ionut Cezar Buciu
- Doctoral School, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania
| | - Eugen Nicolae Tieranu
- Department of Cardiology, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania
- Department of Cardiology, Craiova Emergency Clinical County Hospital, 200642 Craiova, Romania
| | | | - Ovidiu Mircea Zlatian
- Department of Microbiology, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania
- Medical Laboratory, Craiova Emergency Clinical County Hospital, 200642 Craiova, Romania
| | - Ionut Donoiu
- Department of Cardiology, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania
- Department of Cardiology, Craiova Emergency Clinical County Hospital, 200642 Craiova, Romania
| | - Constantin Militaru
- Department of Cardiology, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania
- Department of Cardiology, Craiova Emergency Clinical County Hospital, 200642 Craiova, Romania
- Cardiomed Hospital, 200032 Craiova, Romania
| | - Sebastian Militaru
- Department of Cardiology, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania
- Department of Cardiology, Craiova Emergency Clinical County Hospital, 200642 Craiova, Romania
- Cardiomed Hospital, 200032 Craiova, Romania
| | - Cristian Militaru
- Department of Cardiology, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania
- Department of Cardiology, Craiova Emergency Clinical County Hospital, 200642 Craiova, Romania
- Cardiomed Hospital, 200032 Craiova, Romania
| |
Collapse
|
22
|
Nurmohamed NS, Gaillard EL, Malkasian S, de Groot RJ, Ibrahim S, Bom MJ, Kaiser Y, Earls JP, Min JK, Kroon J, Planken RN, Danad I, van Rosendael AR, Choi AD, Stroes ES, Knaapen P. Lipoprotein(a) and Long-Term Plaque Progression, Low-Density Plaque, and Pericoronary Inflammation. JAMA Cardiol 2024; 9:826-834. [PMID: 39018040 PMCID: PMC11255968 DOI: 10.1001/jamacardio.2024.1874] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Accepted: 05/16/2024] [Indexed: 07/18/2024]
Abstract
Importance Lipoprotein(a) (Lp[a]) is a causal risk factor for cardiovascular disease; however, long-term effects on coronary atherosclerotic plaque phenotype, high-risk plaque formation, and pericoronary adipose tissue inflammation remain unknown. Objective To investigate the association of Lp(a) levels with long-term coronary artery plaque progression, high-risk plaque, and pericoronary adipose tissue inflammation. Design, Setting, and Participants This single-center prospective cohort study included 299 patients with suspected coronary artery disease (CAD) who underwent per-protocol repeated coronary computed tomography angiography (CCTA) imaging with an interscan interval of 10 years. Thirty-two patients were excluded because of coronary artery bypass grafting, resulting in a study population of 267 patients. Data for this study were collected from October 2008 to October 2022 and analyzed from March 2023 to March 2024. Exposures The median scan interval was 10.2 years. Lp(a) was measured at follow-up using an isoform-insensitive assay. CCTA scans were analyzed with a previously validated artificial intelligence-based algorithm (atherosclerosis imaging-quantitative computed tomography). Main Outcome and Measures The association between Lp(a) and change in percent plaque volumes was investigated in linear mixed-effects models adjusted for clinical risk factors. Secondary outcomes were presence of low-density plaque and presence of increased pericoronary adipose tissue attenuation at baseline and follow-up CCTA imaging. Results The 267 included patients had a mean age of 57.1 (SD, 7.3) years and 153 were male (57%). Patients with Lp(a) levels of 125 nmol/L or higher had twice as high percent atheroma volume (6.9% vs 3.0%; P = .01) compared with patients with Lp(a) levels less than 125 nmol/L. Adjusted for other risk factors, every doubling of Lp(a) resulted in an additional 0.32% (95% CI, 0.04-0.60) increment in percent atheroma volume during the 10 years of follow-up. Every doubling of Lp(a) resulted in an odds ratio of 1.23 (95% CI, 1.00-1.51) and 1.21 (95% CI, 1.01-1.45) for the presence of low-density plaque at baseline and follow-up, respectively. Patients with higher Lp(a) levels had increased pericoronary adipose tissue attenuation around both the right coronary artery and left anterior descending at baseline and follow-up. Conclusions and Relevance In this long-term prospective serial CCTA imaging study, higher Lp(a) levels were associated with increased progression of coronary plaque burden and increased presence of low-density noncalcified plaque and pericoronary adipose tissue inflammation. These data suggest an impact of elevated Lp(a) levels on coronary atherogenesis of high-risk, inflammatory, rupture-prone plaques over the long term.
Collapse
Affiliation(s)
- Nick S. Nurmohamed
- Department of Cardiology, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
- Department of Vascular Medicine, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
- Division of Cardiology, The George Washington University School of Medicine, Washington, DC
| | - Emilie L. Gaillard
- Department of Cardiology, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
- Department of Vascular Medicine, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
| | - Shant Malkasian
- Department of Radiological Sciences, Medical Sciences I, University of California, Irvine, California
| | - Robin J. de Groot
- Department of Cardiology, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| | - Shirin Ibrahim
- Department of Vascular Medicine, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
| | - Michiel J. Bom
- Department of Cardiology, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| | - Yannick Kaiser
- Department of Vascular Medicine, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
| | - James P. Earls
- Division of Cardiology, The George Washington University School of Medicine, Washington, DC
- Cleerly, Denver, Colorado
| | | | - Jeffrey Kroon
- Department of Experimental Vascular Medicine, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
- Laboratory of Angiogenesis and Vascular Metabolism, VIB-KU Leuven Center for Cancer Biology, Leuven, Belgium
| | - R. Nils Planken
- Department of Radiology and Nuclear Medicine, Amsterdam UMC, Universiteit van Amsterdam, Amsterdam, the Netherlands
| | - Ibrahim Danad
- Department of Cardiology, University Medical Center Utrecht, Utrecht, the Netherlands
| | | | - Andrew D. Choi
- Division of Cardiology, The George Washington University School of Medicine, Washington, DC
| | - Erik S.G. Stroes
- Department of Vascular Medicine, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
| | - Paul Knaapen
- Department of Cardiology, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| |
Collapse
|
23
|
Sosnowska B, Lewek J, Adach W, Mierczak K, Bielecka-Dąbrowa A, Szosland K, Zygmunt A, Dąbrowski J, Banach M. The prevalence, patients' characteristics, and hyper-Lp(a)-emia risk factors in the Polish population. The first results from the PMMHRI-Lp(a) Registry. Prog Cardiovasc Dis 2024; 86:54-61. [PMID: 39191356 DOI: 10.1016/j.pcad.2024.08.004] [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] [Indexed: 08/29/2024]
Abstract
BACKGROUND The knowledge on the prevalence of elevated lipoprotein(a) (Lp(a)), patients' characteristics, and nongenetic risk factors is scarce in some regions including Poland, the largest Central and Eastern European country. Thus, we aimed to present the results from the Lp(a) registry established in Poland's 2nd largest, supra-regional hospital - the Polish Mother's Memorial Hospital Research Institute (PMMHRI). METHODS The PMMHRI-Lp(a)-Registry was established in January 2022. Since that time all consecutive patients of the Departments of Cardiology, Endocrinology, and outpatient cardiology, diabetology and metabolic clinics have been included. The indications for Lp(a) measurement in the registry are based on the 2021 Polish Lipid Guidelines and new Polish recommendations on the management of elevated Lp(a) (2024). Lp(a) was determined using Sentinel's Lp(a) Ultra, an Immunoturbidimetric quantitative test (Sentinel, Milan, Italy), and the results are presented in mg/dL. RESULTS 511 patients were included in the registry between Jan 2022 and 15th May 2024. The mean age of patients was 48.21 years. Female patients represented 53.42 % of the population. Elevated Lp(a) levels above 30 and 50 mg/dL were detected in 142 (27.79 %), and 101 (19.8 %) patients, respectively. The mean Lp(a) level was 30.45 ± 42.50 mg/dL, with no significant sex differences [mean for men: 28.80 mg/dL; women: 31.89 mg/dL]. There were also no significant differences between those with and without: coronary artery disease (CAD), dyslipidemia, stroke, heart failure, cancer, diabetes, chronic kidney disease, and thyroid disease. The significant Lp(a) level difference was observed in those with a history of myocardial infarction (MI) vs those without (51.47 ± 55.16 vs 28.09 ± 37.51 mg/dL, p < 0.001). However, when we divided those with premature vs no premature MI, no significant difference in Lp(a) level was observed (51.43 ± 57.82 vs 51.52 ± 53.18 mg/dL, p = 0.95). Lipid-lowering therapy (LLT) at baseline did not significantly affect Lp(a) level, with only significant differences for the highest doses of rosuvastatin (p < 0.05) and in those treated with ezetimibe (as a part of the combination therapy; 44.73 ± 54.94 vs 26.84 ± 37.11 mg/dL, p < 0.001). For selected patients (n = 43; 8.42 %) with at least two Lp(a) measurements (mean time distance: 7 ± 5 months, range 1-20 months) we did not observe statistically significant visit-to-visit variability (mean difference: 3.25 mg/dL; r = 0.079, p = 0.616). While dividing the whole population into those with Lp(a) ≤30 mg/dL and > 30 mg/dL, the only hyper-Lp(a)-emia prevalence differences were seen for FH diagnosis (12.88 vs 21.43; p = 0.017), MI prevalence (6.52 vs 16.90 %; p < 0.001), thyroid disease diagnosis (18.14 vs 26.76 %; p = 0.033) and ezetimibe treatment (18.58 vs 30.77 %, p = 0.036). A similar pattern was observed while dividing the whole population on those with Lp(a) ≤50 mg/dL (125 nmol/L) and > 50 mg/dL (125 nmol/L) except for no statistical difference for thyroid disease. CONCLUSIONS These results strongly emphasize that Lp(a) should be measured commonly, as its high level is highly prevalent (even every 3rd patient) in patients at cardiovascular disease (CVD) risk in primary and secondary prevention, requiring risk re-stratification and optimization of the treatment. This is especially important in the regions that characterize baseline high CVD risk, which refers to most CEE countries, including Poland.
Collapse
Affiliation(s)
- Bożena Sosnowska
- Department of Preventive Cardiology and Lipidology, Medical University of Lodz (MUL), Lodz, Poland
| | - Joanna Lewek
- Department of Preventive Cardiology and Lipidology, Medical University of Lodz (MUL), Lodz, Poland; Department of Cardiology and Adult Congenital Heart Diseases, Polish Mother's Memorial Hospital Research Institute (PMMHRI), 93-338 Lodz, Poland
| | - Weronika Adach
- Department of Preventive Cardiology and Lipidology, Medical University of Lodz (MUL), Lodz, Poland
| | - Karina Mierczak
- Department of Preventive Cardiology and Lipidology, Medical University of Lodz (MUL), Lodz, Poland
| | - Agata Bielecka-Dąbrowa
- Department of Preventive Cardiology and Lipidology, Medical University of Lodz (MUL), Lodz, Poland; Department of Endocrinology and Metabolic Diseases, Polish Mother's Memorial Hospital Research Institute, Lodz, Poland
| | - Konrad Szosland
- Department of Endocrinology and Metabolic Diseases, Polish Mother's Memorial Hospital Research Institute, Lodz, Poland
| | - Arkadiusz Zygmunt
- Department of Endocrinology and Metabolic Diseases, Medical University of Lodz, 90-419 Lodz, Poland
| | - Jan Dąbrowski
- Department of Endocrinology and Metabolic Diseases, Polish Mother's Memorial Hospital Research Institute, Lodz, Poland
| | - Maciej Banach
- Department of Preventive Cardiology and Lipidology, Medical University of Lodz (MUL), Lodz, Poland; Department of Cardiology and Adult Congenital Heart Diseases, Polish Mother's Memorial Hospital Research Institute (PMMHRI), 93-338 Lodz, Poland; Cardiovascular Research Centre, University of Zielona Gora, 65-417 Zielona Gora, Poland.
| |
Collapse
|
24
|
Riley TM, Sapp PA, Kris-Etherton PM, Petersen KS. Effects of saturated fatty acid consumption on lipoprotein (a): a systematic review and meta-analysis of randomized controlled trials. Am J Clin Nutr 2024; 120:619-629. [PMID: 38964657 DOI: 10.1016/j.ajcnut.2024.06.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Revised: 06/10/2024] [Accepted: 06/28/2024] [Indexed: 07/06/2024] Open
Abstract
BACKGROUND An inverse relationship between saturated fatty acid (SFA) intake and Lp(a) concentration has been observed; however, there has been no quantification of this effect. OBJECTIVES The objective was to determine whether SFA consumption alters Lp(a) concentrations among adults without atherosclerotic cardiovascular disease (ASCVD). METHODS A systematic review and meta-analysis of randomized controlled trials contrasting a lower SFA diet(s) with a higher SFA diet(s) among adults without ASCVD was conducted. PubMed, Cochrane Central Register of Clinical Trials, clinicaltrials.gov, and Web of Science databases and registers were searched through October 2023. The standardized mean difference (SMD) in Lp(a) between diets lower and higher in SFA [percentage of energy (%E)] was determined using random-effects meta-analysis. Analyses were also conducted to examine the effect of replacing SFA with carbohydrates (CHO), monounsaturated (MUFAs), polyunsaturated (PUFAs), or trans fatty acids (TFAs). RESULTS In total, 6255 publications were identified in the systematic search. Twenty-six publications reporting 27 randomized controlled trials, including 1325 participants and 49 diet comparisons, were included. The mean difference in SFA between lower and higher SFA diets was 7.6%E (3.7%-17.8%E). After lower SFA diets, Lp(a) concentration was higher (SMD: 0.14; 95% confidence interval [CI]: 0.03, 0.24) than after higher SFA diets. Subgroup analyses showed higher Lp(a) following diets where SFA was replaced by CHO (trials = 8; n = 539; SMD: 0.21; 95% CI: 0.02, 0.40) or TFAs (trials = 8; n = 300; SMD: 0.32; 95% CI: 0.17, 0.48). No differences in Lp(a) were observed when MUFA (trials = 16; n = 641; SMD: 0.04; 95% CI: -0.08, 0.16) or PUFA (trials = 8; n = 415; SMD: 0.09; 95% CI: -0.04, 0.22) replaced SFA. CONCLUSIONS Lower SFA diets modestly increase Lp(a) compared to higher SFA diets among individuals without ASCVD. This effect appeared to be driven by replacement of SFA with CHO or TFA. Research investigating the atherogenicity of diet-induced Lp(a) changes is needed to inform dietary management of lipid/lipoprotein disorders. This trial was registered with PROSPERO as CRD42020154169.
Collapse
Affiliation(s)
- Terrence M Riley
- Pennington Biomedical Research Center, Louisiana State University, Baton Rouge, LA, United States; Department of Nutritional Sciences, The Pennsylvania State University, PA, United States.
| | - Philip A Sapp
- Department of Nutritional Sciences, The Pennsylvania State University, PA, United States; Research, Nutrition, and Innovation, Athletic Greens International, Carson City, NV, United States
| | - Penny M Kris-Etherton
- Pennington Biomedical Research Center, Louisiana State University, Baton Rouge, LA, United States
| | - Kristina S Petersen
- Pennington Biomedical Research Center, Louisiana State University, Baton Rouge, LA, United States
| |
Collapse
|
25
|
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
|
26
|
Ratajczak J, Kubica A, Pietrzykowski Ł, Michalski P, Kosobucka-Ozdoba A, Buczkowski K, Krintus M, Jankowski P, Kubica J. Lipoprotein (a) and the Occurrence of Lipid Disorders and Other Cardiovascular Risk Factors in Patients without Diagnosed Cardiovascular Disease. J Clin Med 2024; 13:4649. [PMID: 39200791 PMCID: PMC11354547 DOI: 10.3390/jcm13164649] [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: 07/03/2024] [Revised: 07/29/2024] [Accepted: 08/05/2024] [Indexed: 09/02/2024] Open
Abstract
Background: Elevated lipoprotein (a) [Lp(a)] concentrations are linked mainly to genetic factors. The relationship between Lp(a) and other lipid disorders or cardiovascular (CV) risk factors has been less investigated. The aim of this study was to assess the occurrence of lipid disorders and other CV risk factors according to Lp(a) concentrations. Methods: A cross-sectional analysis of 200 primary-care patients who had not been diagnosed with CV disease was conducted. The following risk factors were assessed: older age, history of hypertension, diabetes mellitus or dyslipidemia, smoking, lack of physical activity, body mass index (BMI), and waist circumference. The following lipid parameters were measured: total cholesterol (TC), low-density lipoprotein cholesterol (LDL-C), non-high-density lipoprotein cholesterol (non-HDL-C), high-density lipoprotein cholesterol (HDL-C), triglycerides (TG), and small, dense LDL (sdLDL-C). Patients were divided into two groups based on their Lp(a) concentrations: <30 mg/dL and ≥30 mg/dL. Results: In 70% of patients, the Lp(a) concentration was <30 mg/dL. The concentrations of lipid parameters did not differ between the groups. The rate of patients with sdLDL-C >1.0 mmol/L was higher in the low-Lp(a) group (10.0 vs. 1.7%, p = 0.04), with no significant differences regarding the other analyzed lipid disorders (p > 0.05). Both in the low- and high-Lp(a) group, most patients had two other abnormal lipid factors (45.0% and 60.0%, respectively). The distribution of impaired lipid parameters (p = 0.41) and other CV risk factors (p = 0.16) was similar in both groups. There was a lower rate of patients >60 years old (15.0% vs. 32.9%, p = 0.01) and with a BMI ≥ 25 kg/m2 (46.7% vs. 63.6%, p = 0.026) in the high-Lp(a) group, and previously diagnosed hyperlipidemia was more prevalent in this group (65.0% vs. 47.1%, p = 0.02). The occurrence of other cardiovascular risk factors did not differ significantly between the Lp(a) groups (p > 0.05). In the high-Lp(a) group, the highest proportion (25.0%) had two CV risk factors, and in the low-Lp(a) group, 31.4% had four CV risk factors. Conclusions: An elevated Lp(a) concentration is not related to the number of conventional CV risk factors or other impairment major lipid parameters.
Collapse
Affiliation(s)
- Jakub Ratajczak
- Department of Cardiac Rehabilitation and Health Promotion, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Torun, 85-094 Bydgoszcz, Poland; (J.R.); (A.K.); (P.M.); (A.K.-O.)
| | - Aldona Kubica
- Department of Cardiac Rehabilitation and Health Promotion, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Torun, 85-094 Bydgoszcz, Poland; (J.R.); (A.K.); (P.M.); (A.K.-O.)
| | - Łukasz Pietrzykowski
- Department of Cardiac Rehabilitation and Health Promotion, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Torun, 85-094 Bydgoszcz, Poland; (J.R.); (A.K.); (P.M.); (A.K.-O.)
| | - Piotr Michalski
- Department of Cardiac Rehabilitation and Health Promotion, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Torun, 85-094 Bydgoszcz, Poland; (J.R.); (A.K.); (P.M.); (A.K.-O.)
| | - Agata Kosobucka-Ozdoba
- Department of Cardiac Rehabilitation and Health Promotion, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Torun, 85-094 Bydgoszcz, Poland; (J.R.); (A.K.); (P.M.); (A.K.-O.)
| | - Krzysztof Buczkowski
- Department of Family Medicine, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Torun, 85-094 Bydgoszcz, Poland;
| | - Magdalena Krintus
- Department of Laboratory Medicine, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Torun, 85-094 Bydgoszcz, Poland;
| | - Piotr Jankowski
- Department of Internal Medicine and Geriatric Cardiology, Center of Postgraduate Medical Education, 01-813 Warsaw, Poland;
- Department of Epidemiology and Health Promotion, School of Public Health, Center of Postgraduate Medical Education, 01-826 Warsaw, Poland
| | - Jacek Kubica
- Department of Cardiology and Internal Medicine, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Torun, 85-094 Bydgoszcz, Poland;
| |
Collapse
|
27
|
Batool H, Khan M, Ain Q, Chughtai OR, Khan MD, Khan MI, Sadiq F. Frequency of lipoprotein(a) testing and its levels in Pakistani population. Glob Cardiol Sci Pract 2024; 2024:e202437. [PMID: 39351483 PMCID: PMC11439424 DOI: 10.21542/gcsp.2024.37] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Accepted: 07/23/2024] [Indexed: 10/04/2024] Open
Abstract
BACKGROUND Lipoprotein(a) [Lp(a)] is a highly atherogenic particle identified as an independent risk factor for the development of atherosclerotic cardiovascular disease (ASCVD). This study aimed to investigate the frequency of Lp(a) testing and the incidence of elevated Lp(a) levels in the Pakistani population. METHODS For this observational study, Lp(a) and lipid profile data from five years (June 2015 to October 2020) were acquired from the electronic patient records of a diagnostic laboratory with a countrywide network. The association of age and total cholesterol (TC), high-density lipoprotein cholesterol (HDL-C), low-density lipoprotein cholesterol (LDL-C), non-HDL, and triglyceride (TG) levels with two thresholds for Lp(a), that is, <30 mg/dL and ≥30 mg/dL, was calculated using the Kruskal-Wallis test, while the association between Lp(a) levels and lipid variables was calculated using Spearman correlation. RESULTS For five years, 1060 tests were conducted, averaging 212 tests per year. Of these, 37.2% showed Lp(a) levels above 30 mg/dL. No significant differences were observed in the results between males and females. However, younger individuals displayed significantly higher Lp(a) levels. Additionally, there was only a weak correlation between the Lp(a) levels and other lipid variables. CONCLUSION Despite being recognized as a risk factor for ASCVD in the Pakistani population, only a small proportion of the large population underwent Lp(a) testing. Moreover, a significant proportion of the population exceeded this threshold.
Collapse
Affiliation(s)
- Hijab Batool
- Chemical Pathology, Chughtai Institute of Pathology, Lahore, Pakistan
| | - Madeeha Khan
- Directorate of Research, Shifa Tameer-e-Millat University, Pitras Bukhari Road, H-8/4, Islamabad 44000, Pakistan
- Atta ur Rehman School of Applied Biosciences, National University of Sciences and Technology, H-12, Islamabad 44000, Pakistan
| | - Quratul Ain
- Directorate of Research, Shifa Tameer-e-Millat University, Pitras Bukhari Road, H-8/4, Islamabad 44000, Pakistan
- Translational Genomics Laboratory, Department of Biosciences, COMSATS University Islamabad, Islamabad, Pakistan
| | - Omar R. Chughtai
- Chemical Pathology, Chughtai Institute of Pathology, Lahore, Pakistan
| | - Muhammad D. Khan
- Chemical Pathology, Chughtai Institute of Pathology, Lahore, Pakistan
| | - Mohammad I. Khan
- Shifa Tameer-e-Millat University, Pitras Bukhari Road, H-8/4, Islamabad 44000, Pakistan
- Department of Vascular Surgery, Shifa International Hospital Pitras Bukhari Road, H-8/4, Islamabad 44000, Pakistan
| | - Fouzia Sadiq
- Directorate of Research, Shifa Tameer-e-Millat University, Pitras Bukhari Road, H-8/4, Islamabad 44000, Pakistan
| |
Collapse
|
28
|
Law HG, Stanhope KL, Zhang W, Myagmarsuren M, Jamshed ZM, Khan MA, Bang H, Havel PJ, Berglund L, Enkhmaa B. Lipoprotein(a) and diet: consuming sugar-sweetened beverages lowers lipoprotein(a) levels in obese and overweight adults. J Lipid Res 2024; 65:100588. [PMID: 38969065 PMCID: PMC11345294 DOI: 10.1016/j.jlr.2024.100588] [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: 04/04/2024] [Revised: 06/12/2024] [Accepted: 06/28/2024] [Indexed: 07/07/2024] Open
Abstract
Lipoprotein(a) [Lp(a)] contributes to cardiovascular disease risk. A genetically determined size polymorphism in apolipoprotein(a) [apo(a)], determined by the number of Kringle (K) repeats, inversely regulates Lp(a) levels. Nongenetic factors including dietary saturated fat influence Lp(a) levels. However, less is known about the effects of carbohydrates including dietary sugars. In this double-blind, parallel arm study among 32 overweight/obese adults, we investigated the effect of consuming glucose- or fructose-sweetened beverages providing 25% of energy requirements for 10 weeks on Lp(a) level and assessed the role of the apo(a) size polymorphism. The mean (±SD) age of participants was 54 ± 8 years, 50% were women, and 75% were of European descent. Following the 10-week intervention, Lp(a) level was reduced by an average (±SEM) of -13.2% ± 4.3% in all participants (P = 0.005); -15.3% ± 7.8% in the 15 participants who consumed glucose (P = 0.07); and -11.3% ± 4.5% in the 17 participants who consumed fructose (P = 0.02), without any significant difference in the effect between the two sugar groups. Relative changes in Lp(a) levels were similar across subgroups of lower versus higher baseline Lp(a) level or carrier versus noncarrier of an atherogenic small (≤22K) apo(a) size. In contrast, LDL-C increased. In conclusion, in older, overweight/obese adults, consuming sugar-sweetened beverages reduced Lp(a) levels by ∼13% independently of apo(a) size variability and the type of sugar consumed. The Lp(a) response was opposite to that of LDL-C and triglyceride concentrations. These findings suggest that metabolic pathways might impact Lp(a) levels.
Collapse
Affiliation(s)
- Hayley G Law
- Department of Internal Medicine, University of California Davis, Davis, CA, USA
| | - Kimber L Stanhope
- Department of Molecular Biosciences, School of Veterinary Medicine and Department of Nutrition, University of California Davis, Davis, CA, USA
| | - Wei Zhang
- Department of Internal Medicine, University of California Davis, Davis, CA, USA
| | | | - Zahraa M Jamshed
- Department of Internal Medicine, University of California Davis, Davis, CA, USA
| | - Muhammad A Khan
- Department of Internal Medicine, University of California Davis, Davis, CA, USA
| | - Heejung Bang
- Department of Public Health Sciences, School of Medicine, University of California Davis, Davis, CA, USA
| | - Peter J Havel
- Department of Molecular Biosciences, School of Veterinary Medicine and Department of Nutrition, University of California Davis, Davis, CA, USA
| | - Lars Berglund
- Department of Internal Medicine, University of California Davis, Davis, CA, USA
| | - Byambaa Enkhmaa
- Department of Internal Medicine, University of California Davis, Davis, CA, USA.
| |
Collapse
|
29
|
Nuotio P, Lankinen MA, Meuronen T, de Mello VD, Sallinen T, Virtanen KA, Pihlajamäki J, Laakso M, Schwab U. Dietary n-3 alpha-linolenic and n-6 linoleic acids modestly lower serum lipoprotein(a) concentration but differentially influence other atherogenic lipoprotein traits: A randomized trial. Atherosclerosis 2024; 395:117562. [PMID: 38714425 DOI: 10.1016/j.atherosclerosis.2024.117562] [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: 11/14/2023] [Revised: 04/17/2024] [Accepted: 04/18/2024] [Indexed: 05/09/2024]
Abstract
BACKGROUND AND AIMS Lipoprotein(a) [Lp(a)] is a causal, genetically determined cardiovascular risk factor. Limited evidence suggests that dietary unsaturated fat may increase serum Lp(a) concentration by 10-15 %. Linoleic acid may increase Lp(a) concentration through its endogenous conversion to arachidonic acid, a process regulated by the fatty acid desaturase (FADS) gene cluster. We aimed to compare the Lp(a) and other lipoprotein trait-modulating effects of dietary alpha-linolenic (ALA) and linoleic acids (LA). Additionally, we examined whether FADS1 rs174550 genotype modifies Lp(a) responses. METHODS A genotype-based randomized trial was performed in 118 men homozygous for FADS1 rs174550 SNP (TT or CC). After a 4-week run-in period, the participants were randomized to 8-week intervention diets enriched with either Camelina sativa oil (ALA diet) or sunflower oil (LA diet) 30-50 mL/day based on their BMI. Serum lipid profile was measured at baseline and at the end of the intervention. RESULTS ALA diet lowered serum Lp(a) concentration by 7.3 % (p = 0.003) and LA diet by 9.5 % (p < 0.001) (p = 0.089 for between-diet difference). Both diets led to greater absolute decreases in individuals with higher baseline Lp(a) concentration (p < 0.001). Concentrations of LDL cholesterol (LDL-C), non-HDL-C, remnant-C, and apolipoprotein B were lowered more by the ALA diet (p < 0.01). Lipid or lipoprotein responses were not modified by the FADS1 rs174550 genotype. CONCLUSIONS A considerable increase in either dietary ALA or LA from vegetable oils has a similar Lp(a)-lowering effect, whereas ALA may lower other major atherogenic lipids and lipoproteins to a greater extent than LA. Genetic differences in endogenous PUFA conversion may not influence serum Lp(a) concentration.
Collapse
Affiliation(s)
- Petrus Nuotio
- Institute of Public Health and Clinical Nutrition, School of Medicine, University of Eastern Finland, 70211, Kuopio, Finland.
| | - Maria A Lankinen
- Institute of Public Health and Clinical Nutrition, School of Medicine, University of Eastern Finland, 70211, Kuopio, Finland
| | - Topi Meuronen
- Food Sciences Unit, Department of Life Technologies, Faculty of Technology, University of Turku, 20500, Turku, Finland
| | - Vanessa D de Mello
- Institute of Public Health and Clinical Nutrition, School of Medicine, University of Eastern Finland, 70211, Kuopio, Finland
| | - Taisa Sallinen
- Institute of Public Health and Clinical Nutrition, School of Medicine, University of Eastern Finland, 70211, Kuopio, Finland
| | - Kirsi A Virtanen
- Institute of Public Health and Clinical Nutrition, School of Medicine, University of Eastern Finland, 70211, Kuopio, Finland; Department of Medicine, Endocrinology and Clinical Nutrition, Kuopio University Hospital, 70029, Kuopio, Finland; Turku PET Centre, University of Turku, 20520, Turku, Finland
| | - Jussi Pihlajamäki
- Institute of Public Health and Clinical Nutrition, School of Medicine, University of Eastern Finland, 70211, Kuopio, Finland; Department of Medicine, Endocrinology and Clinical Nutrition, Kuopio University Hospital, 70029, Kuopio, Finland
| | - Markku Laakso
- Institute of Clinical Medicine, Internal Medicine, University of Eastern Finland, 70029, Kuopio, Finland; Kuopio University Hospital, Kuopio, Finland
| | - Ursula Schwab
- Institute of Public Health and Clinical Nutrition, School of Medicine, University of Eastern Finland, 70211, Kuopio, Finland; Department of Medicine, Endocrinology and Clinical Nutrition, Kuopio University Hospital, 70029, Kuopio, Finland
| |
Collapse
|
30
|
Zimodro JM, Mucha M, Berthold HK, Gouni-Berthold I. Lipoprotein Metabolism, Dyslipidemia, and Lipid-Lowering Therapy in Women: A Comprehensive Review. Pharmaceuticals (Basel) 2024; 17:913. [PMID: 39065763 PMCID: PMC11279947 DOI: 10.3390/ph17070913] [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: 06/20/2024] [Revised: 07/04/2024] [Accepted: 07/08/2024] [Indexed: 07/28/2024] Open
Abstract
Lipid-lowering therapy (LLT) is a cornerstone of atherosclerotic cardiovascular disease prevention. Although LLT might lead to different reductions in low-density lipoprotein cholesterol (LDL-C) levels in women and men, LLT diminishes cardiovascular risk equally effectively in both sexes. Despite similar LLT efficacy, the use of high-intensity statins, ezetimibe, and proprotein convertase subtilisin/kexin type 9 inhibitors is lower in women compared to men. Women achieve the guideline-recommended LDL-C levels less often than men. Greater cholesterol burden is particularly prominent in women with familial hypercholesterolemia. In clinical practice, women and men with dyslipidemia present with different cardiovascular risk profiles and disease manifestations. The concentrations of LDL-C, lipoprotein(a), and other blood lipids differ between women and men over a lifetime. Dissimilar levels of LLT target molecules partially result from sex-specific hormonal and genetic determinants of lipoprotein metabolism. Hence, to evaluate a potential need for sex-specific LLT, this comprehensive review (i) describes the impact of sex on lipoprotein metabolism and lipid profile, (ii) highlights sex differences in cardiovascular risk among patients with dyslipidemia, (iii) presents recent, up-to-date clinical trial and real-world data on LLT efficacy and safety in women, and (iv) discusses the diverse medical needs of women and men with dyslipidemia and increased cardiovascular risk.
Collapse
Affiliation(s)
- Jakub Michal Zimodro
- 1st Chair and Department of Cardiology, Medical University of Warsaw, 02-097 Warsaw, Poland
| | - Magda Mucha
- Faculty of Medicine, Medical University of Bialystok, 15-089 Bialystok, Poland
| | - Heiner K. Berthold
- Department of Internal Medicine and Geriatrics, Bethel Clinic (EvKB), 33611 Bielefeld, Germany
| | - Ioanna Gouni-Berthold
- Center for Endocrinology, Diabetes and Preventive Medicine, Faculty of Medicine, University Hospital Cologne, University of Cologne, Kerpener Str. 62, 50937 Cologne, Germany
| |
Collapse
|
31
|
Saeki Y, Sawaguchi J, Akita S, Takamura TA, Fujibayashi K, Wakasa M, Akao H, Kitayama M, Kawai Y, Kajinami K. Initial decrease in the lipoprotein(a) level is a novel prognostic biomarker in patients with acute coronary syndrome. World J Cardiol 2024; 16:329-338. [PMID: 38993583 PMCID: PMC11235205 DOI: 10.4330/wjc.v16.i6.329] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Revised: 04/22/2024] [Accepted: 04/28/2024] [Indexed: 06/24/2024] Open
Abstract
BACKGROUND Lipoprotein(a) [Lp(a)] is a causal risk factor for atherosclerotic cardiovascular diseases; however, its role in acute coronary syndrome (ACS) remains unclear. AIM To investigate the hypothesis that the Lp(a) levels are altered by various conditions during the acute phase of ACS, resulting in subsequent cardiovascular events. METHODS From September 2009 to May 2016, 377 patients with ACS who underwent emergent coronary angiography, and 249 who completed ≥ 1000 d of follow-up were enrolled. Lp(a) levels were measured using an isoform-independent assay at each time point from before percutaneous coronary intervention (PCI) to 48 h after PCI. The primary endpoint was the occurrence of major adverse cardiac events (MACE; cardiac death, other vascular death, ACS, and non-cardiac vascular events). RESULTS The mean circulating Lp(a) level decreased significantly from pre-PCI (0 h) to 12 h after (19.0 mg/dL to 17.8 mg/dL, P < 0.001), and then increased significantly up to 48 h after (19.3 mg/dL, P < 0.001). The changes from 0 to 12 h [Lp(a)Δ0-12] significantly correlated with the basal levels of creatinine [Spearman's rank correlation coefficient (SRCC): -0.181, P < 0.01] and Lp(a) (SRCC: -0.306, P < 0.05). Among the tertiles classified according to Lp(a)Δ0-12, MACE was significantly more frequent in the lowest Lp(a)Δ0-12 group than in the remaining two tertile groups (66.2% vs 53.6%, P = 0.034). A multivariate analysis revealed that Lp(a)Δ0-12 [hazard ratio (HR): 0.96, 95% confidence interval (95%CI): 0.92-0.99] and basal creatinine (HR: 1.13, 95%CI: 1.05-1.22) were independent determinants of subsequent MACE. CONCLUSION Circulating Lp(a) levels in patients with ACS decreased significantly after emergent PCI, and a greater decrease was independently associated with a worse prognosis.
Collapse
Affiliation(s)
- Yasuhiko Saeki
- Department of Cardiology, Kanazawa Medical University, Uchinada 9200293, Japan
| | - Jun Sawaguchi
- Department of Cardiology, Kanazawa Medical University, Uchinada 9200293, Japan
| | - Satori Akita
- Department of Cardiology, Kanazawa Medical University, Uchinada 9200293, Japan
| | - Taka-Aki Takamura
- Department of Cardiology, Kanazawa Medical University, Uchinada 9200293, Japan
| | - Kosuke Fujibayashi
- Department of Cardiology, Kanazawa Medical University, Uchinada 9200293, Japan
| | - Minoru Wakasa
- Department of Cardiology, Kanazawa Medical University, Uchinada 9200293, Japan
| | - Hironobu Akao
- Department of Cardiology, Kanazawa Medical University, Uchinada 9200293, Japan
| | - Michihiko Kitayama
- Trans-catheter Cardiovascular Therapeutics, Kanazawa Medical University, Uchinada 9200293, Japan
| | - Yasuyuki Kawai
- Department of Cardiology, Kanazawa Medical University, Uchinada 9200293, Japan
| | - Kouji Kajinami
- Department of Cardiology, Kanazawa Medical University, Uchinada 9200293, Japan.
| |
Collapse
|
32
|
Volgman AS, Koschinsky ML, Mehta A, Rosenson RS. Genetics and Pathophysiological Mechanisms of Lipoprotein(a)-Associated Cardiovascular Risk. J Am Heart Assoc 2024; 13:e033654. [PMID: 38879448 PMCID: PMC11255763 DOI: 10.1161/jaha.123.033654] [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] [Indexed: 06/19/2024]
Abstract
Elevated lipoprotein(a) is a genetically transmitted codominant trait that is an independent risk driver for cardiovascular disease. Lipoprotein(a) concentration is heavily influenced by genetic factors, including LPA kringle IV-2 domain size, single-nucleotide polymorphisms, and interleukin-1 genotypes. Apolipoprotein(a) is encoded by the LPA gene and contains 10 subtypes with a variable number of copies of kringle -2, resulting in >40 different apolipoprotein(a) isoform sizes. Genetic loci beyond LPA, such as APOE and APOH, have been shown to impact lipoprotein(a) levels. Lipoprotein(a) concentrations are generally 5% to 10% higher in women than men, and there is up to a 3-fold difference in median lipoprotein(a) concentrations between racial and ethnic populations. Nongenetic factors, including menopause, diet, and renal function, may also impact lipoprotein(a) concentration. Lipoprotein(a) levels are also influenced by inflammation since the LPA promoter contains an interleukin-6 response element; interleukin-6 released during the inflammatory response results in transient increases in plasma lipoprotein(a) levels. Screening can identify elevated lipoprotein(a) levels and facilitate intensive risk factor management. Several investigational, RNA-targeted agents have shown promising lipoprotein(a)-lowering effects in clinical studies, and large-scale lipoprotein(a) testing will be fundamental to identifying eligible patients should these agents become available. Lipoprotein(a) testing requires routine, nonfasting blood draws, making it convenient for patients. Herein, we discuss the genetic determinants of lipoprotein(a) levels, explore the pathophysiological mechanisms underlying the association between lipoprotein(a) and cardiovascular disease, and provide practical guidance for lipoprotein(a) testing.
Collapse
Affiliation(s)
| | - Marlys L. Koschinsky
- Robarts Research Institute, Schulich School of Medicine and DentistryWestern UniversityLondonONCanada
| | | | - Robert S. Rosenson
- Metabolism and Lipids Program, Mount Sinai Fuster Heart HospitalIcahn School of Medicine at Mount SinaiNew YorkNYUSA
| |
Collapse
|
33
|
Masson W, Barbagelata L, Godinez-Leiva E, Genua I, Nogueira JP. Association between hepatic steatosis and lipoprotein(a) levels in non-alcoholic patients: A systematic review. Indian J Gastroenterol 2024; 43:536-547. [PMID: 38036914 DOI: 10.1007/s12664-023-01457-2] [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: 04/19/2023] [Accepted: 09/02/2023] [Indexed: 12/02/2023]
Abstract
BACKGROUND AND OBJECTIVES It is well known that lipid abnormalities exist in the context of non-alcoholic fatty liver disease (NAFLD). The association between lipoprotein(a) [Lp(a)] levels and NAFLD is poorly understood. The main objective of the present study was to assess the association between Lp(a) levels and NAFLD. METHODS This systematic review was performed according to Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guidelines (PROSPERO CRD42023392526). A literature search was performed to detect studies that evaluated the association between Lp(a) levels, NAFLD and steatohepatitis (NASH). RESULTS Ten observational studies, including 40,045 patients, were identified and considered eligible for this systematic review. There were 9266 subjects in the NAFLD groups and 30,779 individuals in the respective control groups. Five studies evaluated patients with NAFLD (hepatic steatosis was associated with lower Lp(a) levels in four studies, while the remaining showed opposite results). Two studies evaluating NASH patients showed that Lp(a) levels were not different compared to controls. However, the increment of Lp(a) levels was correlated with liver fibrosis in one of them. In addition, one study analyzed simultaneously patients with NAFLD and NASH, showing a neutral result in NAFLD patients and a positive relationship in NASH patients. Two studies that included patients with the new definition of metabolic-associated fatty liver disease (MAFLD) also showed neutral results. CONCLUSION Although there could be an association between Lp(a) levels and hepatic steatosis, the results of the studies published to date are contradictory and not definitive.
Collapse
Affiliation(s)
- Walter Masson
- Servicio de Cardiología, Hospital Italiano de Buenos Aires, Perón 4190, C1199ABB, Ciudad Autónoma de Buenos Aires, Argentina
| | - Leandro Barbagelata
- Servicio de Cardiología, Hospital Italiano de Buenos Aires, Perón 4190, C1199ABB, Ciudad Autónoma de Buenos Aires, Argentina.
| | - Eddison Godinez-Leiva
- Division of Endocrinology, Diabetes and Metabolism, University of Florida, Gainesville, FL, USA
- Universidad Internacional de Las Américas, San José, Costa Rica
| | - Idoia Genua
- Endocrinology and Nutrition Department, Hospital de La Santa Creu I Sant Pau, Autonomous University of Barcelona (UAB), Barcelona, Spain
- Institut d'Investigació Biomèdica Sant Pau (IIB SANT PAU), Barcelona, Spain
| | - Juan Patricio Nogueira
- Centro de Investigación en Endocrinología, Nutrición Y Metabolismo (CIENM), Facultad de Ciencias de La Salud, Universidad Nacional de Formosa, Formosa, Argentina
| |
Collapse
|
34
|
Cignarella A, Bolego C, Barton M. Sex and sex steroids as determinants of cardiovascular risk. Steroids 2024; 206:109423. [PMID: 38631602 DOI: 10.1016/j.steroids.2024.109423] [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: 12/21/2023] [Revised: 04/08/2024] [Accepted: 04/14/2024] [Indexed: 04/19/2024]
Abstract
There are considerable sex differences regarding the risk of cardiovascular disease (CVD), including arterial hypertension, coronary artery disease (CAD) and stroke, as well as chronic renal disease. Women are largely protected from these conditions prior to menopause, and the risk increases following cessation of endogenous estrogen production or after surgical menopause. Cardiovascular diseases in women generally begin to occur at a later age than in men (on average with a delay of 10 years). Cessation of estrogen production also impacts metabolism, increasing the risk of developing obesity and diabetes. In middle-aged individuals, hypertension develops earlier and faster in women than in men, and smoking increases cardiovascular risk to a greater degree in women than it does in men. It is not only estrogen that affects female cardiovascular health and plays a protective role until menopause: other sex hormones such as progesterone and androgen hormones generate a complex balance that differentiates heart and blood vessel function in women compared to men. Estrogens improve vasodilation of epicardial coronary arteries and the coronary microvasculature by augmenting the release of vasodilating factors such as nitric oxide and prostacyclin, which are mechanisms of coronary vasodilatation that are more pronounced in women compared to men. Estrogens are also powerful inhibitors of inflammation, which in part explains their protective effects on CVD and chronic renal disease. Emerging evidence suggests that sex chromosomes also play a significant role in shaping cardiovascular risk. The cardiovascular protection conferred by endogenous estrogens may be extended by hormone therapy, especially using bioidentical hormones and starting treatment early after menopause.
Collapse
Affiliation(s)
| | - Chiara Bolego
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Padova, Italy
| | - Matthias Barton
- Molecular Internal Medicine, University of Zürich, Zürich, Switzerland; Andreas Grüntzig Foundation, Zürich, Switzerland.
| |
Collapse
|
35
|
Pavlatos N, Kalra DK. The Role of Lipoprotein(a) in Peripheral Artery Disease. Biomedicines 2024; 12:1229. [PMID: 38927436 PMCID: PMC11200468 DOI: 10.3390/biomedicines12061229] [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: 05/05/2024] [Revised: 05/25/2024] [Accepted: 05/29/2024] [Indexed: 06/28/2024] Open
Abstract
Lipoprotein(a) is a low-density-lipoprotein-like particle that consists of apolipoprotein(a) bound to apolipoprotein(b). It has emerged as an established causal risk factor for atherosclerotic cardiovascular disease, stroke, and aortic valve stenosis through multifactorial pathogenic mechanisms that include inflammation, atherogenesis, and thrombosis. Despite an estimated 20% of the global population having elevated lipoprotein(a) levels, testing remains underutilized due to poor awareness and a historical lack of effective and safe therapies. Although lipoprotein(a) has a strong association with coronary artery disease and cerebrovascular disease, its relationship with peripheral artery disease is less well established. In this article, we review the epidemiology, biology, and pathogenesis of lipoprotein(a) as it relates to peripheral artery disease. We also discuss emerging treatment options to help mitigate major adverse cardiac and limb events in this population.
Collapse
Affiliation(s)
- Nicholas Pavlatos
- Department of Internal Medicine, University of Louisville School of Medicine, Louisville, KY 40202, USA;
| | - Dinesh K. Kalra
- Division of Cardiology, University of Louisville School of Medicine, Louisville, KY 40202, USA
| |
Collapse
|
36
|
Kaur G, Abdelrahman K, Berman AN, Biery DW, Shiyovich A, Huck D, Garshick M, Blankstein R, Weber B. Lipoprotein(a): Emerging insights and therapeutics. Am J Prev Cardiol 2024; 18:100641. [PMID: 38646022 PMCID: PMC11033089 DOI: 10.1016/j.ajpc.2024.100641] [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: 11/16/2023] [Revised: 02/08/2024] [Accepted: 02/24/2024] [Indexed: 04/23/2024] Open
Abstract
The strong association between lipoprotein (a) [Lp(a)] and atherosclerotic cardiovascular disease has led to considerations of Lp(a) being a potential target for mitigating residual cardiovascular risk. While approximately 20 % of the population has an Lp(a) level greater than 50 mg/dL, there are no currently available pharmacological lipid-lowering therapies that have demonstrated substantial reduction in Lp(a). Novel therapies to lower Lp(a) include antisense oligonucleotides and small-interfering ribonucleic acid molecules and have shown promising results in phase 2 trials. Phase 3 trials are currently underway and will test the causal relationship between Lp(a) and ASCVD and whether lowering Lp(a) reduces cardiovascular outcomes. In this review, we summarize emerging insights related to Lp(a)'s role as a risk-enhancing factor for ASCVD, association with calcific aortic stenosis, effects of existing therapies on Lp(a) levels, and variations amongst patient populations. The evolving therapeutic landscape of emerging therapeutics is further discussed.
Collapse
Affiliation(s)
- Gurleen Kaur
- Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | | | - Adam N. Berman
- Division of Cardiovascular Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - David W. Biery
- Division of Cardiovascular Medicine, Brigham and Women's Hospital, Boston, MA, USA
- Albert Einstein College of Medicine, New York, NY, USA
| | - Arthur Shiyovich
- Division of Cardiovascular Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Daniel Huck
- Division of Cardiovascular Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | | | - Ron Blankstein
- Division of Cardiovascular Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Brittany Weber
- Division of Cardiovascular Medicine, Brigham and Women's Hospital, Boston, MA, USA
| |
Collapse
|
37
|
Reyes-Soffer G, Yeang C, Michos ED, Boatwright W, Ballantyne CM. High lipoprotein(a): Actionable strategies for risk assessment and mitigation. Am J Prev Cardiol 2024; 18:100651. [PMID: 38646021 PMCID: PMC11031736 DOI: 10.1016/j.ajpc.2024.100651] [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: 12/08/2023] [Revised: 03/13/2024] [Accepted: 03/17/2024] [Indexed: 04/23/2024] Open
Abstract
High levels of lipoprotein(a) [Lp(a)] are causal for atherosclerotic cardiovascular disease (ASCVD). Lp(a) is the most prevalent inherited dyslipidemia and strongest genetic ASCVD risk factor. This risk persists in the presence of at target, guideline-recommended, LDL-C levels and adherence to lifestyle modifications. Epidemiological and genetic evidence supporting its causal role in ASCVD and calcific aortic stenosis continues to accumulate, although various facets regarding Lp(a) biology (genetics, pathophysiology, and expression across race/ethnic groups) are not yet fully understood. The evolving nature of clinical guidelines and consensus statements recommending universal measurements of Lp(a) and the scientific data supporting its role in multiple disease states reinforce the clinical merit to start population screening for Lp(a) now. There is a current gap in the implementation of recommendations for primary and secondary cardiovascular disease (CVD) prevention in those with high Lp(a), in part due to a lack of protocols for management strategies. Importantly, targeted apolipoprotein(a) [apo(a)]-lowering therapies that reduce Lp(a) levels in patients with high Lp(a) are in phase 3 clinical development. This review focuses on the identification and clinical management of patients with high Lp(a). Specifically, we highlight the clinical value of measuring Lp(a) and its use in determining Lp(a)-associated CVD risk by providing actionable guidance, based on scientific knowledge, that can be utilized now to mitigate risk caused by high Lp(a).
Collapse
Affiliation(s)
| | - Calvin Yeang
- Department of Medicine, UC San Diego Health, CA, USA
| | - Erin D Michos
- Division of Cardiology, Johns Hopkins University School of Medicine, MD, USA
| | | | | |
Collapse
|
38
|
Koschinsky ML, Bajaj A, Boffa MB, Dixon DL, Ferdinand KC, Gidding SS, Gill EA, Jacobson TA, Michos ED, Safarova MS, Soffer DE, Taub PR, Wilkinson MJ, Wilson DP, Ballantyne CM. A focused update to the 2019 NLA scientific statement on use of lipoprotein(a) in clinical practice. J Clin Lipidol 2024; 18:e308-e319. [PMID: 38565461 DOI: 10.1016/j.jacl.2024.03.001] [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: 03/04/2024] [Accepted: 03/06/2024] [Indexed: 04/04/2024]
Abstract
Since the 2019 National Lipid Association (NLA) Scientific Statement on Use of Lipoprotein(a) in Clinical Practice was issued, accumulating epidemiological data have clarified the relationship between lipoprotein(a) [Lp(a)] level and cardiovascular disease risk and risk reduction. Therefore, the NLA developed this focused update to guide clinicians in applying this emerging evidence in clinical practice. We now have sufficient evidence to support the recommendation to measure Lp(a) levels at least once in every adult for risk stratification. Individuals with Lp(a) levels <75 nmol/L (30 mg/dL) are considered low risk, individuals with Lp(a) levels ≥125 nmol/L (50 mg/dL) are considered high risk, and individuals with Lp(a) levels between 75 and 125 nmol/L (30-50 mg/dL) are at intermediate risk. Cascade screening of first-degree relatives of patients with elevated Lp(a) can identify additional individuals at risk who require intervention. Patients with elevated Lp(a) should receive early, more-intensive risk factor management, including lifestyle modification and lipid-lowering drug therapy in high-risk individuals, primarily to reduce low-density lipoprotein cholesterol (LDL-C) levels. The U.S. Food and Drug Administration approved an indication for lipoprotein apheresis (which reduces both Lp(a) and LDL-C) in high-risk patients with familial hypercholesterolemia and documented coronary or peripheral artery disease whose Lp(a) level remains ≥60 mg/dL [∼150 nmol/L)] and LDL-C ≥ 100 mg/dL on maximally tolerated lipid-lowering therapy. Although Lp(a) is an established independent causal risk factor for cardiovascular disease, and despite the high prevalence of Lp(a) elevation (∼1 of 5 individuals), measurement rates are low, warranting improved screening strategies for cardiovascular disease prevention.
Collapse
Affiliation(s)
- Marlys L Koschinsky
- Department of Physiology & Pharmacology and Robarts Research Institute, Schulich School of Medicine & Dentistry, Western University, London, Ontario, Canada (Drs Koschinsky, Boffa)
| | - Archna Bajaj
- Department of Medicine, University of Pennsylvania, Philadelphia, PA, USA (Drs Bajaj, Soffer)
| | - Michael B Boffa
- Department of Physiology & Pharmacology and Robarts Research Institute, Schulich School of Medicine & Dentistry, Western University, London, Ontario, Canada (Drs Koschinsky, Boffa)
| | - Dave L Dixon
- Department of Pharmacotherapy & Outcomes Science, Virginia Commonwealth University School of Pharmacy, Richmond, VA, USA (Dr Dixon)
| | - Keith C Ferdinand
- Department of Medicine, Tulane University School of Medicine, New Orleans, LA, USA (Dr. Ferdinand)
| | - Samuel S Gidding
- Department of Genomic Health, Geisinger. Danville, PA, USA (Dr Gidding)
| | - Edward A Gill
- Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA (Dr Gill)
| | - Terry A Jacobson
- Department of Medicine, Emory University School of Medicine, Atlanta, GA, USA (Dr Jacobson)
| | - Erin D Michos
- Division of Cardiology, Johns Hopkins University School of Medicine, Baltimore, MD, USA (Dr Michos)
| | - Maya S Safarova
- Division of Cardiovascular Medicine, Department of Internal Medicine, Froedtert & the Medical College of Wisconsin, Milwaukee, WI, USA (Dr Safarova)
| | - Daniel E Soffer
- Department of Medicine, University of Pennsylvania, Philadelphia, PA, USA (Drs Bajaj, Soffer)
| | - Pam R Taub
- Department of Medicine, University of California San Diego, La Jolla, CA, USA (Drs Taub, Wilkinson)
| | - Michael J Wilkinson
- Department of Medicine, University of California San Diego, La Jolla, CA, USA (Drs Taub, Wilkinson)
| | - Don P Wilson
- Department of Pediatric Endocrinology and Diabetes, Cook Children's Medical Center, Fort Worth, TX, USA (Dr Wilson)
| | - Christie M Ballantyne
- Department of Medicine, Baylor College of Medicine, Houston, TX, USA (Dr Ballantyne).
| |
Collapse
|
39
|
Bess C, Mehta A, Joshi PH. All we need to know about lipoprotein(a). Prog Cardiovasc Dis 2024; 84:27-33. [PMID: 38759878 DOI: 10.1016/j.pcad.2024.05.007] [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: 05/14/2024] [Accepted: 05/14/2024] [Indexed: 05/19/2024]
Abstract
Lipoprotein(a) [Lp(a)], a genetically determined macromolecular complex, is independently and causally associated with atherosclerotic cardiovascular disease (ASCVD) and calcific aortic stenosis via proposed proinflammatory, prothrombotic, and proatherogenic mechanisms. While Lp(a) measurement standardization issues are being resolved, several guidelines now support testing Lp(a) at least once in each adult's lifetime for ASCVD risk prediction which can foster implementation of more aggressive primary or secondary prevention therapies. Currently, there are several emerging targeted Lp(a) lowering therapies in active clinical investigation for safety and cardiovascular benefit among both primary and secondary prevention populations. First degree relatives of patients with high Lp(a) should be encouraged to undergo cascade screening. Primary prevention patients with high Lp(a) should consider obtaining a coronary calcium score for further risk estimation and to guide further ASCVD risk factor management including consideration of low dose aspirin therapy. Secondary prevention patients with high Lp(a) levels should consider adding PCSK9 inhibition to statin therapy.
Collapse
Affiliation(s)
- Courtney Bess
- Department of Internal Medicine, Division of Cardiology, University of Texas Southwestern, Dallas, TX, United States of America; Parkland Health and Hospital System, Dallas, TX, United States of America
| | - Anurag Mehta
- VCU Health Pauley Health Center, Richmond, VA, United States of America
| | - Parag H Joshi
- Department of Internal Medicine, Division of Cardiology, University of Texas Southwestern, Dallas, TX, United States of America; Parkland Health and Hospital System, Dallas, TX, United States of America.
| |
Collapse
|
40
|
Botana López MA. Lipoprotein (a): Is its systematic determination indicated? ENDOCRINOL DIAB NUTR 2024; 71:191-193. [PMID: 38834503 DOI: 10.1016/j.endien.2024.02.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Accepted: 02/28/2024] [Indexed: 06/06/2024]
|
41
|
Abstract
Prolonged or excessive exposure to oxidized phospholipids (OxPLs) generates chronic inflammation. OxPLs are present in atherosclerotic lesions and can be detected in plasma on apolipoprotein B (apoB)-containing lipoproteins. When initially conceptualized, OxPL-apoB measurement in plasma was expected to reflect the concentration of minimally oxidized LDL, but, surprisingly, it correlated more strongly with plasma lipoprotein(a) (Lp(a)) levels. Indeed, experimental and clinical studies show that Lp(a) particles carry the largest fraction of OxPLs among apoB-containing lipoproteins. Plasma OxPL-apoB levels provide diagnostic information on the presence and extent of atherosclerosis and improve the prognostication of peripheral artery disease and first and recurrent myocardial infarction and stroke. The addition of OxPL-apoB measurements to traditional cardiovascular risk factors improves risk reclassification, particularly in patients in intermediate risk categories, for whom improving decision-making is most impactful. Moreover, plasma OxPL-apoB levels predict cardiovascular events with similar or greater accuracy than plasma Lp(a) levels, probably because this measurement reflects both the genetics of elevated Lp(a) levels and the generalized or localized oxidation that modifies apoB-containing lipoproteins and leads to inflammation. Plasma OxPL-apoB levels are reduced by Lp(a)-lowering therapy with antisense oligonucleotides and by lipoprotein apheresis, niacin therapy and bariatric surgery. In this Review, we discuss the role of role OxPLs in the pathophysiology of atherosclerosis and Lp(a) atherogenicity, and the use of OxPL-apoB measurement for improving prognosis, risk reclassification and therapeutic interventions.
Collapse
Affiliation(s)
- Sotirios Tsimikas
- Division of Cardiovascular Medicine, University of California San Diego, La Jolla, CA, USA.
| | - Joseph L Witztum
- Division of Endocrinology and Metabolism, University of California San Diego, La Jolla, CA, USA
| |
Collapse
|
42
|
Kielb J, Saffak S, Weber J, Baensch L, Shahjerdi K, Celik A, Farahat N, Riek S, Chavez-Talavera O, Grandoch M, Polzin A, Kelm M, Dannenberg L. Transformation or replacement - Effects of hormone therapy on cardiovascular risk. Pharmacol Ther 2024; 254:108592. [PMID: 38286163 DOI: 10.1016/j.pharmthera.2024.108592] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Revised: 12/28/2023] [Accepted: 12/29/2023] [Indexed: 01/31/2024]
Abstract
Hormone therapy (HT) is important and frequently used both regarding replacement therapy (HRT) and gender affirming therapy (GAHT). While HRT has been effective in addressing symptoms related to hormone shortage, several side effects have been described. In this context, there are some studies that show increased cardiovascular risk. However, there are also studies reporting protective aspects of HT. Nevertheless, the exact impact of HT on cardiovascular risk and the underlying mechanisms remain poorly understood. This article explores the relationship between diverse types of HT and cardiovascular risk, focusing on mechanistic insights of the underlying hormones on platelet and leukocyte function as well as on effects on endothelial and adipose tissue cells.
Collapse
Affiliation(s)
- Julia Kielb
- Department of Cardiology, Pulmonology, and Vascular Medicine, Medical Faculty of the Heinrich Heine University Düsseldorf, Düsseldorf, Germany; Cardiovascular Research Institute Düsseldorf (CARID), Germany
| | - Süreyya Saffak
- Department of Cardiology, Pulmonology, and Vascular Medicine, Medical Faculty of the Heinrich Heine University Düsseldorf, Düsseldorf, Germany; Cardiovascular Research Institute Düsseldorf (CARID), Germany
| | - Jessica Weber
- Department of Cardiology, Pulmonology, and Vascular Medicine, Medical Faculty of the Heinrich Heine University Düsseldorf, Düsseldorf, Germany; Cardiovascular Research Institute Düsseldorf (CARID), Germany
| | - Leonard Baensch
- Department of Cardiology, Pulmonology, and Vascular Medicine, Medical Faculty of the Heinrich Heine University Düsseldorf, Düsseldorf, Germany; Cardiovascular Research Institute Düsseldorf (CARID), Germany
| | - Khatereh Shahjerdi
- Department of Cardiology, Pulmonology, and Vascular Medicine, Medical Faculty of the Heinrich Heine University Düsseldorf, Düsseldorf, Germany; Cardiovascular Research Institute Düsseldorf (CARID), Germany
| | - Aylin Celik
- Department of Cardiology, Pulmonology, and Vascular Medicine, Medical Faculty of the Heinrich Heine University Düsseldorf, Düsseldorf, Germany; Cardiovascular Research Institute Düsseldorf (CARID), Germany
| | - Nora Farahat
- Department of Cardiology, Pulmonology, and Vascular Medicine, Medical Faculty of the Heinrich Heine University Düsseldorf, Düsseldorf, Germany; Cardiovascular Research Institute Düsseldorf (CARID), Germany
| | - Sally Riek
- Department of Cardiology, Pulmonology, and Vascular Medicine, Medical Faculty of the Heinrich Heine University Düsseldorf, Düsseldorf, Germany; Cardiovascular Research Institute Düsseldorf (CARID), Germany
| | - Oscar Chavez-Talavera
- Department of Cardiology, Pulmonology, and Vascular Medicine, Medical Faculty of the Heinrich Heine University Düsseldorf, Düsseldorf, Germany; Cardiovascular Research Institute Düsseldorf (CARID), Germany
| | - Maria Grandoch
- Institute for Translational Pharmacology, Medical Faculty and University Hospital of Düsseldorf, Heinrich-Heine University Düsseldorf, Düsseldorf, Germany
| | - Amin Polzin
- Department of Cardiology, Pulmonology, and Vascular Medicine, Medical Faculty of the Heinrich Heine University Düsseldorf, Düsseldorf, Germany; Cardiovascular Research Institute Düsseldorf (CARID), Germany
| | - Malte Kelm
- Department of Cardiology, Pulmonology, and Vascular Medicine, Medical Faculty of the Heinrich Heine University Düsseldorf, Düsseldorf, Germany; Cardiovascular Research Institute Düsseldorf (CARID), Germany
| | - Lisa Dannenberg
- Department of Cardiology, Pulmonology, and Vascular Medicine, Medical Faculty of the Heinrich Heine University Düsseldorf, Düsseldorf, Germany; Cardiovascular Research Institute Düsseldorf (CARID), Germany.
| |
Collapse
|
43
|
Sosnowska B, Stepinska J, Mitkowski P, Bielecka-Dabrowa A, Bobrowska B, Budzianowski J, Burchardt P, Chlebus K, Dobrowolski P, Gasior M, Jankowski P, Kubica J, Mickiewicz A, Mysliwiec M, Osadnik T, Prejbisz A, Rajtar-Salwa R, Wita K, Witkowski A, Gil R, Banach M. Recommendations of the Experts of the Polish Cardiac Society (PCS) and the Polish Lipid Association (PoLA) on the diagnosis and management of elevated lipoprotein(a) levels. Arch Med Sci 2024; 20:8-27. [PMID: 38414479 PMCID: PMC10895977 DOI: 10.5114/aoms/183522] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Accepted: 01/31/2024] [Indexed: 02/29/2024] Open
Abstract
Lipoprotein(a) [Lp(a)] is made up of a low-density lipoprotein (LDL) particle and a specific apolipoprotein(a). The blood concentration of Lp(a) is approximately 90% genetically determined, and the main genetic factor determining Lp(a) levels is the size of the apo(a) isoform, which is determined by the number of KIV2 domain repeats. The size of the apo(a) isoform is inversely proportional to the blood concentration of Lp(a). Lp(a) is a strong and independent cardiovascular risk factor. Elevated Lp(a) levels ≥ 50 mg/dl (≥ 125 nmol/l) are estimated to occur in more than 1.5 billion people worldwide. However, determination of Lp(a) levels is performed far too rarely, including Poland, where, in fact, it is only since the 2021 guidelines of the Polish Lipid Association (PoLA) and five other scientific societies that Lp(a) measurements have begun to be performed. Determination of Lp(a) concentrations is not easy due to, among other things, the different sizes of the apo(a) isoforms; however, the currently available certified tests make it possible to distinguish between people with low and high cardiovascular risk with a high degree of precision. In 2022, the first guidelines for the management of patients with elevated lipoprotein(a) levels were published by the European Atherosclerosis Society (EAS) and the American Heart Association (AHA). The first Polish guidelines are the result of the work of experts from the two scientific societies and their aim is to provide clear, practical recommendations for the determination and management of elevated Lp(a) levels.
Collapse
Affiliation(s)
- Bożena Sosnowska
- Department of Preventive Cardiology and Lipidology, Medical University of Lodz (MUL), Lodz, Poland
| | | | - Przemyslaw Mitkowski
- 1 Department of Cardiology, Poznan University of Medical Sciences, Poznan, Poland
| | - Agata Bielecka-Dabrowa
- Department of Preventive Cardiology and Lipidology, Medical University of Lodz (MUL), Lodz, Poland
- Department of Cardiology and Adult Congenital Defects, Polish Mother’s Memorial Hospital Research Institute (PMMHRI), Lodz, Poland
| | - Beata Bobrowska
- Department of Clinical Cardiology and Cardiovascular Interventions, University Hospital in Krakow, Krakow, Poland
| | - Jan Budzianowski
- Department of Interventional Cardiology and Cardiac Surgery, University of Zielona Gora, Collegium Medicum, Zielona Gora, Poland
- Multidisciplinary Hospital, Nowa Sol, Poland
| | - Pawel Burchardt
- Department of Cardiology, J. Strus Hospital, Poznan, Poland
- Department of Hypertension, Angiology and Internal Medicine, Poznan University of Medical Sciences, Poznan, Poland
| | - Krzysztof Chlebus
- National Center for Familial Hypercholesterolemia, 1 Chair and Department of Cardiology, Medical University of Gdansk, Gdansk, Poland
| | - Piotr Dobrowolski
- Department of Epidemiology, Cardiovascular Disease Prevention and Health Promotion, National Institute of Cardiology, Warsaw, Poland
| | - Mariusz Gasior
- 3 Department of Cardiology, Silesian Centre for Heart Diseases, Faculty of Medical Sciences in Zabrze, Medical University of Silesia, Zabrze, Poland
| | - Piotr Jankowski
- Department of Internal Medicine and Geriatric Cardiology, Medical Centre for Postgraduate Education, Warsaw, Poland
| | - Jacek Kubica
- Department of Cardiology and Internal Medicine, Collegium Medicum, Nicolaus Copernicus University, Bydgoszcz, Poland
| | - Agnieszka Mickiewicz
- Lipoprotein Apheresis Laboratory, 1 Department of Cardiology, Medical University of Gdansk, Gdansk, Poland
| | - Malgorzata Mysliwiec
- Department of Paediatrics, Diabetology and Endocrinology, Medical University of Gdansk, Gdansk, Poland
| | - Tadeusz Osadnik
- Department of Pharmacology, Faculty of Medical Sciences in Zabrze, Medical University of Silesia, Katowice, Poland
| | - Aleksander Prejbisz
- Department of Epidemiology, Cardiovascular Disease Prevention and Health Promotion, National Institute of Cardiology, Warsaw, Poland
| | - Renata Rajtar-Salwa
- Department of Clinical Cardiology and Cardiovascular Interventions, University Hospital in Krakow, Krakow, Poland
| | - Kristian Wita
- 1 Department of Cardiology, School of Medicine in Katowice, Medical University of Silesia, Katowice, Poland
| | - Adam Witkowski
- Department of Interventional Cardiology and Angiology, National Institute of Cardiology, Warsaw, Poland
| | - Robert Gil
- Department of Cardiology, National Medical Institute of the Ministry of Internal Affairs and Administration, Warsaw, Poland
| | - Maciej Banach
- Department of Preventive Cardiology and Lipidology, Medical University of Lodz (MUL), Lodz, Poland
- Department of Cardiology and Adult Congenital Defects, Polish Mother’s Memorial Hospital Research Institute (PMMHRI), Lodz, Poland
- Ciccarone Center for the Prevention of Cardiovascular Disease, Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| |
Collapse
|
44
|
Tsamoulis D, Kosmas CE, Rallidis LS. Is inverse association between lipoprotein(a) and diabetes mellitus another paradox in cardiometabolic medicine? Expert Rev Endocrinol Metab 2024; 19:63-70. [PMID: 38078437 DOI: 10.1080/17446651.2023.2293108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/17/2023] [Accepted: 12/06/2023] [Indexed: 01/03/2024]
Abstract
INTRODUCTION The impact of Type II Diabetes mellitus (T2DM) on cardiovascular disease (CVD) is well-established, while lipoprotein(a) [Lp(a)] has recently emerged as a recognized CVD risk factor. The rising prevalence of T2DM resulting from modern lifestyles and the development of specific Lp(a)-lowering agents brought the association between T2DM and Lp(a) in the forefront. AREAS COVERED Despite advancements in T2DM treatment, diabetic patients remain at very-high risk of CVD. Lp(a) may, to some extent, contribute to the persistent CVD risk seen in diabetic patients, and the coexistence of T2DM and elevated Lp(a) levels appears to synergistically amplify overall CVD risk. The relationship between T2DM and Lp(a) is paradoxical. On one hand, high Lp(a) plasma concentrations elevate the risk of diabetic microvascular and macrovascular complications. On the other hand, low Lp(a) plasma concentrations have been linked to an increased risk of developing T2DM. EXPERT OPINION Comprehending the association between T2DM and Lp(a) is critical due to the pivotal roles both entities play in overall CVD risk, as well as the unique aspects of their relationship. The mechanisms underlying the inverse association between T2DM and Lp(a) remain incompletely understood, necessitating further meticulous research.
Collapse
Affiliation(s)
- Donatos Tsamoulis
- Second Department of Cardiology, National & Kapodistrian University of Athens, Athens, Greece
| | - Constantine E Kosmas
- Division of Cardiology, Department of Medicine, Montefiore Medical Center, Bronx, NY, USA
| | - Loukianos S Rallidis
- Second Department of Cardiology, National & Kapodistrian University of Athens, Athens, Greece
| |
Collapse
|
45
|
Chen S, Li Z, Li H, Zeng X, Yuan H, Li Y. Novel lipid biomarkers and ratios as risk predictors for premature coronary artery disease: A retrospective analysis of 2952 patients. J Clin Hypertens (Greenwich) 2023; 25:1172-1184. [PMID: 37986641 PMCID: PMC10710552 DOI: 10.1111/jch.14751] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Revised: 10/23/2023] [Accepted: 10/30/2023] [Indexed: 11/22/2023]
Abstract
This study examined the associations between emerging lipid biomarkers (small dense low-density lipoprotein cholesterol [sdLDL-C), lipoprotein(a) [Lp(a)], and free fatty acids [FFA]), two ratios (sdLDL-C/LDL-C and the triglyceride-glucose [TyG) index), and the Gensini score (GS) in patients with premature coronary artery disease (PCAD) in relation to the extent of coronary stenosis. The authors evaluated a cohort of 2952 individuals undergoing coronary angiography (CAG), encompassing those with PCAD (n = 1749), late-onset coronary artery disease (LCAD; n = 328), and non-coronary artery disease (non-CAD; n = 575). Noteworthy differences were observed in the levels of the novel lipid biomarkers and ratio indexes among the PCAD, LCAD, and non-CAD groups (p < .05). Multiple logistic regression analyses pinpointed Lp(a) (OR = 2.62, 95% CI 1.22-5.63, p = .014) and the TyG index (OR = 2.53, 95% CI 1.08-5.93, p = .033) as independent risk factors for PCAD. Furthermore, these biomarkers and ratio indexes discerned substantial distinctions among PCAD patients with varying GS (p < .05). Consequently, these markers can proficiently anticipate the gravity of coronary artery stenosis (GS > 40) in PCAD patients, as evidenced by the ROC analysis. In conclusion, sdLDL-C, Lp(a), FFA, and the sdLDL-C/LDL-C and TyG indexes have considerable potential as risk and diagnostic markers for coronary artery stenosis in individuals afflicted with PCAD.
Collapse
Affiliation(s)
- Si Chen
- Department of Clinical LaboratoryPeking Union Medical College HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
- State Key Laboratory of ComplexSevere and Rare DiseasesPeking Union Medical College HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
- Department of Clinical LaboratoryBeijing Anzhen HospitalCapital Medical UniversityBeijingChina
| | - Zhan Li
- Department of Clinical LaboratoryPeking Union Medical College HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
- State Key Laboratory of ComplexSevere and Rare DiseasesPeking Union Medical College HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Haolong Li
- Department of Clinical LaboratoryPeking Union Medical College HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
- State Key Laboratory of ComplexSevere and Rare DiseasesPeking Union Medical College HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Xiaoli Zeng
- Department of Clinical LaboratoryBeijing Anzhen HospitalCapital Medical UniversityBeijingChina
| | - Hui Yuan
- Department of Clinical LaboratoryBeijing Anzhen HospitalCapital Medical UniversityBeijingChina
| | - Yongzhe Li
- Department of Clinical LaboratoryPeking Union Medical College HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
- State Key Laboratory of ComplexSevere and Rare DiseasesPeking Union Medical College HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| |
Collapse
|
46
|
Pasławska A, Tomasik PJ. Lipoprotein(a)-60 Years Later-What Do We Know? Cells 2023; 12:2472. [PMID: 37887316 PMCID: PMC10605347 DOI: 10.3390/cells12202472] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Revised: 09/17/2023] [Accepted: 10/14/2023] [Indexed: 10/28/2023] Open
Abstract
Lipoprotein(a) (Lp(a)) molecule includes two protein components: apolipoprotein(a) and apoB100. The molecule is the main transporter of oxidized phospholipids (OxPL) in plasma. The concentration of this strongly atherogenic lipoprotein is predominantly regulated by the LPA gene expression. Lp(a) is regarded as a risk factor for several cardiovascular diseases. Numerous epidemiological, clinical and in vitro studies showed a strong association between increased Lp(a) and atherosclerotic cardiovascular disease (ASCVD), calcific aortic valve disease/aortic stenosis (CAVD/AS), stroke, heart failure or peripheral arterial disease (PAD). Although there are acknowledged contributions of Lp(a) to the mentioned diseases, clinicians struggle with many inconveniences such as a lack of well-established treatment lowering Lp(a), and common guidelines for diagnosing or assessing cardiovascular risk among both adult and pediatric patients. Lp(a) levels are different with regard to a particular race or ethnicity and might fluctuate during childhood. Furthermore, the lack of standardization of assays is an additional impediment. The review presents the recent knowledge on Lp(a) based on clinical and scientific research, but also highlights relevant aspects of future study directions that would approach more suitable and effective managing risk associated with increased Lp(a), as well as control the Lp(a) levels.
Collapse
Affiliation(s)
- Anna Pasławska
- Tuchow Health Center, Medical Hospital Laboratory, Szpitalna St. 1, 33-170 Tuchow, Poland;
| | - Przemysław J. Tomasik
- Department of Clinical Biochemistry, Pediatric Institute, College of Medicine, Jagiellonian University, Wielicka St. 265, 30-663 Cracow, Poland
| |
Collapse
|
47
|
Bhatia HS. Aspirin and lipoprotein(a) in primary prevention. Curr Opin Lipidol 2023; 34:214-220. [PMID: 37527183 PMCID: PMC11216950 DOI: 10.1097/mol.0000000000000891] [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] [Indexed: 08/03/2023]
Abstract
PURPOSE OF REVIEW Lipoprotein(a) [Lp(a)] is causally associated with cardiovascular diseases, and elevated levels are highly prevalent. However, there is a lack of available therapies to address Lp(a)-mediated risk. Though aspirin has progressively fallen out of favor for primary prevention, individuals with high Lp(a) may represent a high-risk group that derives a net benefit. RECENT FINDINGS Aspirin has been demonstrated to have a clear benefit in secondary prevention of cardiovascular disease, but recent primary prevention trials have at best demonstrated a small benefit. However, individuals with elevated Lp(a) may be of high risk enough to benefit, particularly given interactions between Lp(a) and the fibrinolytic system / platelets, and the lack of available targeted medical therapies. In secondary analyses of the Women's Health Study (WHS) and the Aspirin in Reducing Events in the Elderly (ASPREE) trial, aspirin use was associated with a significant reduction in cardiovascular events in carriers of genetic polymorphisms associated with elevated Lp(a) levels. Further studies are needed, however, as these studies focused on narrower subsets of the overall population and genetic markers. SUMMARY Individuals with elevated Lp(a) may benefit from aspirin therapy in primary prevention, but further study with plasma Lp(a) levels, broader populations, and randomization of aspirin are needed.
Collapse
Affiliation(s)
- Harpreet S Bhatia
- Division of Cardiovascular Medicine, Department of Medicine, University of California San Diego, La Jolla, California, USA
| |
Collapse
|
48
|
Vinci P, Di Girolamo FG, Panizon E, Tosoni LM, Cerrato C, Pellicori F, Altamura N, Pirulli A, Zaccari M, Biasinutto C, Roni C, Fiotti N, Schincariol P, Mangogna A, Biolo G. Lipoprotein(a) as a Risk Factor for Cardiovascular Diseases: Pathophysiology and Treatment Perspectives. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2023; 20:6721. [PMID: 37754581 PMCID: PMC10531345 DOI: 10.3390/ijerph20186721] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Revised: 07/31/2023] [Accepted: 08/09/2023] [Indexed: 09/28/2023]
Abstract
Cardiovascular disease (CVD) is still a leading cause of morbidity and mortality, despite all the progress achieved as regards to both prevention and treatment. Having high levels of lipoprotein(a) [Lp(a)] is a risk factor for cardiovascular disease that operates independently. It can increase the risk of developing cardiovascular disease even when LDL cholesterol (LDL-C) levels are within the recommended range, which is referred to as residual cardiovascular risk. Lp(a) is an LDL-like particle present in human plasma, in which a large plasminogen-like glycoprotein, apolipoprotein(a) [Apo(a)], is covalently bound to Apo B100 via one disulfide bridge. Apo(a) contains one plasminogen-like kringle V structure, a variable number of plasminogen-like kringle IV structures (types 1-10), and one inactive protease region. There is a large inter-individual variation of plasma concentrations of Lp(a), mainly ascribable to genetic variants in the Lp(a) gene: in the general po-pulation, Lp(a) levels can range from <1 mg/dL to >1000 mg/dL. Concentrations also vary between different ethnicities. Lp(a) has been established as one of the risk factors that play an important role in the development of atherosclerotic plaque. Indeed, high concentrations of Lp(a) have been related to a greater risk of ischemic CVD, aortic valve stenosis, and heart failure. The threshold value has been set at 50 mg/dL, but the risk may increase already at levels above 30 mg/dL. Although there is a well-established and strong link between high Lp(a) levels and coronary as well as cerebrovascular disease, the evidence regarding incident peripheral arterial disease and carotid atherosclerosis is not as conclusive. Because lifestyle changes and standard lipid-lowering treatments, such as statins, niacin, and cholesteryl ester transfer protein inhibitors, are not highly effective in reducing Lp(a) levels, there is increased interest in developing new drugs that can address this issue. PCSK9 inhibitors seem to be capable of reducing Lp(a) levels by 25-30%. Mipomersen decreases Lp(a) levels by 25-40%, but its use is burdened with important side effects. At the current time, the most effective and tolerated treatment for patients with a high Lp(a) plasma level is apheresis, while antisense oligonucleotides, small interfering RNAs, and microRNAs, which reduce Lp(a) levels by targeting RNA molecules and regulating gene expression as well as protein production levels, are the most widely explored and promising perspectives. The aim of this review is to provide an update on the current state of the art with regard to Lp(a) pathophysiological mechanisms, focusing on the most effective strategies for lowering Lp(a), including new emerging alternative therapies. The purpose of this manuscript is to improve the management of hyperlipoproteinemia(a) in order to achieve better control of the residual cardiovascular risk, which remains unacceptably high.
Collapse
Affiliation(s)
- Pierandrea Vinci
- Clinica Medica, Cattinara Hospital, Department of Medical Surgical and Health Science, University of Trieste, 34149 Trieste, Italy; (F.G.D.G.); (E.P.); (L.M.T.); (C.C.); (F.P.); (N.A.); (A.P.); (M.Z.); (N.F.); (G.B.)
| | - Filippo Giorgio Di Girolamo
- Clinica Medica, Cattinara Hospital, Department of Medical Surgical and Health Science, University of Trieste, 34149 Trieste, Italy; (F.G.D.G.); (E.P.); (L.M.T.); (C.C.); (F.P.); (N.A.); (A.P.); (M.Z.); (N.F.); (G.B.)
- SC Assistenza Farmaceutica, Cattinara Hospital, Azienda Sanitaria Universitaria Integrata di Trieste, 34149 Trieste, Italy; (C.B.); (C.R.); (P.S.)
| | - Emiliano Panizon
- Clinica Medica, Cattinara Hospital, Department of Medical Surgical and Health Science, University of Trieste, 34149 Trieste, Italy; (F.G.D.G.); (E.P.); (L.M.T.); (C.C.); (F.P.); (N.A.); (A.P.); (M.Z.); (N.F.); (G.B.)
| | - Letizia Maria Tosoni
- Clinica Medica, Cattinara Hospital, Department of Medical Surgical and Health Science, University of Trieste, 34149 Trieste, Italy; (F.G.D.G.); (E.P.); (L.M.T.); (C.C.); (F.P.); (N.A.); (A.P.); (M.Z.); (N.F.); (G.B.)
| | - Carla Cerrato
- Clinica Medica, Cattinara Hospital, Department of Medical Surgical and Health Science, University of Trieste, 34149 Trieste, Italy; (F.G.D.G.); (E.P.); (L.M.T.); (C.C.); (F.P.); (N.A.); (A.P.); (M.Z.); (N.F.); (G.B.)
| | - Federica Pellicori
- Clinica Medica, Cattinara Hospital, Department of Medical Surgical and Health Science, University of Trieste, 34149 Trieste, Italy; (F.G.D.G.); (E.P.); (L.M.T.); (C.C.); (F.P.); (N.A.); (A.P.); (M.Z.); (N.F.); (G.B.)
| | - Nicola Altamura
- Clinica Medica, Cattinara Hospital, Department of Medical Surgical and Health Science, University of Trieste, 34149 Trieste, Italy; (F.G.D.G.); (E.P.); (L.M.T.); (C.C.); (F.P.); (N.A.); (A.P.); (M.Z.); (N.F.); (G.B.)
| | - Alessia Pirulli
- Clinica Medica, Cattinara Hospital, Department of Medical Surgical and Health Science, University of Trieste, 34149 Trieste, Italy; (F.G.D.G.); (E.P.); (L.M.T.); (C.C.); (F.P.); (N.A.); (A.P.); (M.Z.); (N.F.); (G.B.)
| | - Michele Zaccari
- Clinica Medica, Cattinara Hospital, Department of Medical Surgical and Health Science, University of Trieste, 34149 Trieste, Italy; (F.G.D.G.); (E.P.); (L.M.T.); (C.C.); (F.P.); (N.A.); (A.P.); (M.Z.); (N.F.); (G.B.)
| | - Chiara Biasinutto
- SC Assistenza Farmaceutica, Cattinara Hospital, Azienda Sanitaria Universitaria Integrata di Trieste, 34149 Trieste, Italy; (C.B.); (C.R.); (P.S.)
| | - Chiara Roni
- SC Assistenza Farmaceutica, Cattinara Hospital, Azienda Sanitaria Universitaria Integrata di Trieste, 34149 Trieste, Italy; (C.B.); (C.R.); (P.S.)
| | - Nicola Fiotti
- Clinica Medica, Cattinara Hospital, Department of Medical Surgical and Health Science, University of Trieste, 34149 Trieste, Italy; (F.G.D.G.); (E.P.); (L.M.T.); (C.C.); (F.P.); (N.A.); (A.P.); (M.Z.); (N.F.); (G.B.)
| | - Paolo Schincariol
- SC Assistenza Farmaceutica, Cattinara Hospital, Azienda Sanitaria Universitaria Integrata di Trieste, 34149 Trieste, Italy; (C.B.); (C.R.); (P.S.)
| | - Alessandro Mangogna
- Institute for Maternal and Child Health, I.R.C.C.S “Burlo Garofolo”, 34137 Trieste, Italy;
| | - Gianni Biolo
- Clinica Medica, Cattinara Hospital, Department of Medical Surgical and Health Science, University of Trieste, 34149 Trieste, Italy; (F.G.D.G.); (E.P.); (L.M.T.); (C.C.); (F.P.); (N.A.); (A.P.); (M.Z.); (N.F.); (G.B.)
| |
Collapse
|
49
|
Law HG, Khan MA, Zhang W, Bang H, Rood J, Most M, Lefevre M, Berglund L, Enkhmaa B. Reducing saturated fat intake lowers LDL-C but increases Lp(a) levels in African Americans: the GET-READI feeding trial. J Lipid Res 2023; 64:100420. [PMID: 37482217 PMCID: PMC10445453 DOI: 10.1016/j.jlr.2023.100420] [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: 04/18/2023] [Revised: 07/17/2023] [Accepted: 07/19/2023] [Indexed: 07/25/2023] Open
Abstract
Reducing dietary saturated fatty acids (SFA) intake results in a clinically significant lowering of low-density lipoprotein cholesterol (LDL-C) across ethnicities. In contrast, dietary SFA's role in modulating emerging cardiovascular risk factors in different ethnicities remains poorly understood. Elevated levels of lipoprotein(a) [Lp(a)], an independent cardiovascular risk factor, disproportionally affect individuals of African descent. Here, we assessed the responses in Lp(a) levels to dietary SFA reduction in 166 African Americans enrolled in GET-READI (The Gene-Environment Trial on Response in African Americans to Dietary Intervention), a randomized controlled feeding trial. Participants were fed two diets in random order for 5 weeks each: 1) an average American diet (AAD) (37% total fat: 16% SFA), and 2) a diet similar to the Dietary Approaches to Stop Hypertension (DASH) diet (25% total fat: 6% SFA). The participants' mean age was 35 years, 70% were women, the mean BMI was 28 kg/m2, and the mean LDL-C was 116 mg/dl. Compared to the AAD diet, LDL-C was reduced by the DASH-type diet (mean change: -12 mg/dl) as were total cholesterol (-16 mg/dl), HDL-C (-5 mg/dl), apoA-1 (-9 mg/dl) and apoB-100 (-5 mg/dl) (all P < 0.0001). In contrast, Lp(a) levels increased following the DASH-type diet compared with AAD (median: 58 vs. 44 mg/dl, P < 0.0001). In conclusion, in a large cohort of African Americans, reductions in SFA intake significantly increased Lp(a) levels while reducing LDL-C. Future studies are warranted to elucidate the mechanism(s) underlying the SFA reduction-induced increase in Lp(a) levels and its role in cardiovascular risk across populations.
Collapse
Affiliation(s)
- Hayley G Law
- Department of Internal Medicine, School of Medicine, University of California Davis, Davis, CA, USA
| | - Muhammad A Khan
- Department of Internal Medicine, School of Medicine, University of California Davis, Davis, CA, USA
| | - Wei Zhang
- Department of Internal Medicine, School of Medicine, University of California Davis, Davis, CA, USA
| | - Heejung Bang
- Department of Public Health Sciences, School of Medicine, University of California Davis, Davis, CA, USA
| | - Jennifer Rood
- Pennington Biomedical Research Center, Baton Rouge, LA, USA
| | - Marlene Most
- Pennington Biomedical Research Center, Baton Rouge, LA, USA
| | - Michael Lefevre
- Pennington Biomedical Research Center, Baton Rouge, LA, USA; Department of Nutrition, Utah State University, Logan, UT, USA
| | - Lars Berglund
- Department of Internal Medicine, School of Medicine, University of California Davis, Davis, CA, USA
| | - Byambaa Enkhmaa
- Department of Internal Medicine, School of Medicine, University of California Davis, Davis, CA, USA; Center for Precision Medicine and Data Sciences, School of Medicine, University of California Davis, Davis, CA, USA.
| |
Collapse
|
50
|
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
|