1
|
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 2024:64953. [PMID: 39370270 DOI: 10.5551/jat.64953] [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: 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
|
2
|
Zubirán R, Cruz-Bautista I, Aguilar-Salinas CA. Interaction Between Primary Hyperlipidemias and Type 2 Diabetes: Therapeutic Implications. Diabetes Ther 2024; 15:1979-2000. [PMID: 39080218 PMCID: PMC11330433 DOI: 10.1007/s13300-024-01626-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Accepted: 07/10/2024] [Indexed: 08/18/2024] Open
Abstract
There is a gap of knowledge about the clinical and pathophysiological implications resulting from the interaction between primary hyperlipidemias and type 2 diabetes (T2D). Most of the existing evidence comes from sub-analyses of cohorts; scant information derives from randomized clinical trials. The expected clinical implications of T2D in patients with primary hyperlipidemias is an escalation of their already high cardiovascular risk. There is a need to accurately identify patients with this dual burden and to adequately prescribe lipid-lowering therapies, with the current advancements in newer therapeutic options. This review provides an update on the interactions of primary hyperlipidemias, such as familial combined hyperlipidemia, familial hypercholesterolemia, multifactorial chylomicronemia, lipoprotein (a), and type 2 diabetes.
Collapse
Affiliation(s)
- Rafael Zubirán
- Lipoprotein Metabolism Laboratory, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Ivette Cruz-Bautista
- Unidad de Investigación de Enfermedades Metabólicas, Instituto Nacional de Ciencias Medicas y Nutricion Salvador Zubiran, Mexico City, Mexico
| | - Carlos A Aguilar-Salinas
- Unidad de Investigación de Enfermedades Metabólicas, Instituto Nacional de Ciencias Medicas y Nutricion Salvador Zubiran, Mexico City, Mexico.
- Dirección de Investigación, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico.
- Tecnológico de Monterrey, Escuela de Medicina y Ciencias de la Salud, Mexico City, Mexico.
| |
Collapse
|
3
|
Dyrbuś K, Mędrala Z, Konsek K, Nowowiejska-Wiewióra A, Trzeciak P, Skrzypek M, Cieśla D, Banach M, Gąsior M. Lipoprotein(a) and its impact on cardiovascular disease - the Polish perspective: design and first results of the Zabrze-Lipoprotein(a) Registry. Arch Med Sci 2024; 20:1069-1076. [PMID: 39439677 PMCID: PMC11493035 DOI: 10.5114/aoms/188294] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Accepted: 05/05/2024] [Indexed: 10/25/2024] Open
Abstract
Introduction Lipoprotein(a) [Lp(a)] is an independent risk factor for atherosclerotic cardiovascular disease (ASCVD). Increased Lp(a) concentration > 30 mg/dl (75 nmol/l) and especially >50 mg/dl (125 nmol/l) may cause faster atherosclerosis, being an important and underdiagnosed residual cardiovascular risk factor. Thus, there is a need to characterize further the clinical phenotypes in patients at risk for ASCVD with high Lp(a) levels now and during follow-up, while also looking for the possible impact of geographical differences. Material and methods The Zabrze Lipoprotein(a) Registry (Zabrze-Lip(a)R) was founded on the basis of data from 2,001 consecutive patients with very high cardiovascular risk treated in a tertiary hospital. The registry patients will be followed for at least 5 years with the possibility of extending this period as an open label study. All-cause and cause-specific mortality, hospitalizations, and cardiovascular events, such as myocardial infarction (MI) and stroke, will be assessed. Results The mean age of patients was 66.4 years (females 37.1%). The median Lp(a) concentration in the entire population was 6.6 mg/dl (16.5 nmol/l) (mean: 14.3 ±19.4 mg/dl). 540 (27%) patients had elevated Lp(a) levels above 30 mg/dl (75 nmol/l); they were significantly older (68.8 vs. 66.3 years; p = 0.04), had significantly lower hemoglobin and hematocrit, and higher platelet count and levels of NT-proBNP and C-reactive protein. The prevalence of elevated Lp(a) > 30 mg/dl (75 nmol/l) concentrations was very high in patients with a chronic coronary syndrome (CCS) (52.2% (282/540) vs. 41.5% (607/1461); p < 0.001), in patients undergoing PCI during hospitalization (23.9 vs. 19%; p = 0.01), and in patients with previous MI (20.6% vs. 14.9%; p = 0.0022). In the multivariable analysis, the independent predictors of elevated Lp(a) > 30 mg/dl (75 nmol/l) were only lower Hb values (OR = 0.925; 95% CI: 0.874-0.978; p = 0.006) and higher platelet count (1.002; 95%CI: 1.000-1.003; p < 0.02). Conclusions In Poland, the largest representative of Central and Eastern European countries, 27% of patients at very high cardiovascular risk with established ASCVD experience additional risk related to an elevated Lp(a) level, with every second patient having CCS. Interestingly, only two factors were significantly related to elevated Lp(a) levels: lower Hb values and higher platelet count. However, the clinical relevance of these results needs confirmation.
Collapse
Affiliation(s)
- Krzysztof Dyrbuś
- Silesian Center for Heart Diseases (SCHD), 3 Department of Cardiology, SMDZ in Zabrze, Medical University of Silesia in Katowice, Poland
| | - Zofia Mędrala
- Silesian Center for Heart Diseases (SCHD), 3 Department of Cardiology, SMDZ in Zabrze, Medical University of Silesia in Katowice, Poland
| | - Karolina Konsek
- Silesian Center for Heart Diseases (SCHD), 3 Department of Cardiology, SMDZ in Zabrze, Medical University of Silesia in Katowice, Poland
| | - Alicja Nowowiejska-Wiewióra
- Silesian Center for Heart Diseases (SCHD), 3 Department of Cardiology, SMDZ in Zabrze, Medical University of Silesia in Katowice, Poland
| | - Przemysław Trzeciak
- Silesian Center for Heart Diseases (SCHD), 3 Department of Cardiology, SMDZ in Zabrze, Medical University of Silesia in Katowice, Poland
| | - Michał Skrzypek
- Department of Environmental Health, School of Public Health in Bytom, Medical University of Silesia, Katowice, Poland
| | - Daniel Cieśla
- Silesian Center for Heart Diseases (SCHD), 3 Department of Cardiology, SMDZ in Zabrze, Medical University of Silesia in Katowice, Poland
| | - Maciej Banach
- Department of Preventive Cardiology and Lipidology, Medical University of Lodz, Lodz, Poland
- Ciccarone Center for the Prevention of Cardiovascular Disease, School of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Mariusz Gąsior
- Silesian Center for Heart Diseases (SCHD), 3 Department of Cardiology, SMDZ in Zabrze, Medical University of Silesia in Katowice, Poland
| |
Collapse
|
4
|
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
|
5
|
Jawi M, Al Mahdi HB, Daghistani H, Almoghrabi Y, Algahtani R, Tawati D, Algethami MR, Noorelahi Y, Awan Z. Lipoprotein(a): Levels and Reference Intervals Among People in Saudi Arabia. Diabetes Metab Syndr Obes 2024; 17:2935-2942. [PMID: 39114527 PMCID: PMC11303827 DOI: 10.2147/dmso.s465885] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Accepted: 07/25/2024] [Indexed: 08/10/2024] Open
Abstract
Purpose Blood Lp(a) concentration is recognized as an independent risk factor for cardiovascular disease (CVD). Population-based lipoprotein(a) (Lp[a]) research in Saudi Arabia is rare. Thus, the primary goal of this pilot study was to identify age- and sex-specific reference ranges for Lp(a) levels, in addition to the associations between Lp(a) levels and other atherosclerotic markers in Saudi individuals. Patients and methods A five-year retrospective study of Lp(a) and lipid markers in Saudi patients was conducted using the Al-Borg diagnostics database (2015-2020). The population sample consisted of 361 Saudi individuals aged 18-93 years (162 males, 199 females). An immunoturbidimetric technique was used to determine Lp(a) concentration. Results The mean and median Lp(a) levels in the study population were 35 nmol/L and 50 nmol/L, respectively. Sex and age did not influence Lp(a) values. Lp(a) values showed a minor correlation with other atherosclerotic markers when the Pearson correlation coefficient was used. In Saudi Arabia, the distribution of Lp(a) concentrations is skewed to the left, favoring lower values. Conclusion Lp(a) levels in individuals residing in Saudi Arabia were comparable to those observed in other ethnic groups. Additionally, standardizing Lp(a) measurements according to sex and age may enhance broader applicability and facilitate comparisons across different populations. However, larger studies are required to provide more comprehensive data for comparison.
Collapse
Affiliation(s)
- Motasim Jawi
- Department of Physiology, College of Medicine, University of Jeddah, Jeddah, Saudi Arabia
| | - Hadiah Bassam Al Mahdi
- Al Borg Medical Laboratories, Al Borg Diagnostics, Research and Development Unit, Jeddah, Saudi Arabia
- Department of Biological Sciences, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Hussam Daghistani
- Department of Clinical Biochemistry, Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
- Regenerative Medicine Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Yousef Almoghrabi
- Department of Clinical Biochemistry, Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
- Regenerative Medicine Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Rami Algahtani
- Department of Medicine, College of Medicine, Umm Al-Qura University, Makkah, Saudi Arabia
| | - Dalal Tawati
- Department of Obstetrics and Gynecology, King Abdullah Medical Complex, Jeddah, Saudi Arabia
| | - Mohammed R Algethami
- Department of Family and Community Medicine, College of Medicine, University of Jeddah, Jeddah, Saudi Arabia
| | - Yasser Noorelahi
- Department of Radiology, Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Zuhier Awan
- Department of Clinical Biochemistry, Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
| |
Collapse
|
6
|
Leiter LA, Raal FJ, Schwartz GG, Koenig W, Ray KK, Landmesser U, Han J, Conde LG, Wright RS. Inclisiran in individuals with diabetes or obesity: Post hoc pooled analyses of the ORION-9, ORION-10 and ORION-11 Phase 3 randomized trials. Diabetes Obes Metab 2024; 26:3223-3237. [PMID: 38757725 DOI: 10.1111/dom.15650] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Revised: 04/26/2024] [Accepted: 04/26/2024] [Indexed: 05/18/2024]
Abstract
AIMS To conduct a pooled analysis of Phase 3 trials investigating the efficacy and safety of inclisiran across glycaemic and body mass index (BMI) strata. MATERIALS AND METHODS Participants were randomized 1:1 to receive 300 mg inclisiran sodium or placebo twice yearly, after initial and 3-month doses up to 18 months, with background oral lipid-lowering therapy. Analyses were stratified by glycaemic status (normoglycaemia, prediabetes, and diabetes) or BMI (<25, ≥25 to <30, ≥30 to <35, and ≥35 kg/m2). Co-primary endpoints were percentage and time-adjusted percentage change in low-density lipoprotein (LDL) cholesterol from baseline. Safety was also assessed. RESULTS Baseline characteristics were balanced between treatment arms and across strata. Percent LDL cholesterol change (placebo-corrected) with inclisiran from baseline to Day 510 ranged from -47.6% to -51.9% and from -48.8% to -54.4% across glycaemic/BMI strata, respectively. Similarly, time-adjusted percentage changes after Day 90 and up to Day 540 ranged from -46.8% to -52.0% and from -48.6% to -53.3% across glycaemic/BMI strata, respectively. Inclisiran led to significant reductions in proprotein convertase subtilisin/kexin type 9 and other atherogenic lipids and lipoproteins versus placebo across the glycaemic/BMI strata. The proportions of individuals achieving LDL cholesterol thresholds of <1.8 mmol/L and <1.4 mmol/L with inclisiran increased with increasing glycaemic and BMI strata. Across the glycaemic/BMI strata, a higher proportion of individuals had mild/moderate treatment-emergent adverse events (TEAEs) at the injection site with inclisiran (2.8%-7.7%) versus placebo (0.2%-2.1%). CONCLUSION Inclisiran provided substantial and sustained LDL cholesterol lowering across glycaemic/BMI strata, with a modest excess of transient mild-to-moderate TEAEs at the injection site.
Collapse
Affiliation(s)
- Lawrence A Leiter
- Li Ka Shing Knowledge Institute, St. Michael's Hospital, University of Toronto, Toronto, Canada
| | - Frederick J Raal
- Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Gregory G Schwartz
- Division of Cardiology, University of Colorado School of Medicine, Aurora, Colorado, USA
| | - Wolfgang Koenig
- Deutsches Herzzentrum München, Technische Universität München, Munich, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Munich Heart Alliance, Munich, Germany
- Institute of Epidemiology and Medical Biometry, University of Ulm, Ulm, Germany
| | - Kausik K Ray
- Imperial Centre for Cardiovascular Disease Prevention, Department of Primary Care and Public Health, Imperial College, London, UK
| | - Ulf Landmesser
- Department of Cardiology, Angiology and Intensive Care Medicine, Deutsches Herzzentrum der Charité, Charité University Medicine Berlin, Friede Springer Cardiovascular Prevention Center od Charité, Berlin Institute of Health, DZHK, Partner Site Berlin, Berlin, Germany
| | - Jackie Han
- Novartis Pharmaceuticals Corporation, East Hanover, New Jersey, USA
| | | | - R Scott Wright
- Division of Preventive Cardiology and Department of Cardiology, Mayo Clinic, Rochester, Minnesota, USA
| |
Collapse
|
7
|
Li Y, Kronenberg F, Coassin S, Vardarajan B, Reyes-Soffer G. Ancestry specific distribution of LPA Kringle IV-Type-2 genetic variants highlight associations to apo(a) copy number, glucose, and hypertension. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.07.09.24310176. [PMID: 39040175 PMCID: PMC11261928 DOI: 10.1101/2024.07.09.24310176] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/24/2024]
Abstract
Background High Lp(a) levels contribute to atherosclerotic cardiovascular disease and are tightly regulated by the LPA gene . Lp(a) levels have an inverse correlation with LPA Kringle IV Type-2 (KIV-2) copy number (CN). Black (B) and Hispanic (H) individuals exhibit higher levels of Lp(a), and rates of CVD compared to non-Hispanic Whites (NHW). Therefore, we investigated genetic variations in the LPA KIV-2 region across three ancestries and their associations with metabolic risk factors. Methods Using published pipelines, we analyzed a multi-ethnic whole exome dataset comprising 3,817 participants from the Washington Heights and Inwood Columbia Aging Project (WHICAP): 886 [NHW (23%), 1,811 Caribbean (C) H (47%), and 1,120 B individuals (29%). Rare and common variants (alternative allele carrier frequency, CF < 0.01 or > 0.99 and 0.01 < CF < 0.99, respectively) were identified and KIV-2 CN estimated. The associations of variants and CN with history of heart disease, hypertension (HTN), stroke, lipid levels and clinical diagnosis of Alzheimer's disease (AD) was assessed. A small pilot provided in-silico validation of study findings. Results We report 1421 variants in the LPA KIV-2 repeat region, comprising 267 exonic and 1154 intronic variants. 61.4% of the exonic variants have not been previously described. Three novel exonic variants significantly increase the risk of HTN across all ethnic groups: 4785-C/A (frequency = 78%, odds ratio [OR] = 1.45, p = 0.032), 727-T/C (frequency = 96%, OR = 2.11, p = 0.032), and 723-A/G (frequency = 96%, OR = 1.97, p = 0.038). Additionally, six intronic variants showed associations with HTN: 166-G/A, 387-G/C, 402-G/A, 4527-A/T, 4541-G/A, and 4653-A/T. One intronic variant, 412-C/T, was associated with decreased blood glucose levels (frequency = 72%, β = -14.52, p = 0.02).Three of the associations were not affected after adjusting for LPA KIV-2 CN: 412-C/T (β = -14.2, p = 0.03), 166-G/A (OR = 1.41, p = 0.05), and 387-G/C (OR = 1.40, p = 0.05). KIV CN itself was significantly associated with 314 variants and was negatively correlated with plasma total cholesterol levels. Conclusions In three ancestry groups, we identify novel rare and common LPA KIV-2 region variants. We report new associations of variants with HTN and Glucose levels. These results underscore the genetic complexity of the LPA KIV-2 region in influencing cardiovascular and metabolic health, suggesting potential genetic regulation of pathways that can be studied for research and therapeutic interventions.
Collapse
Affiliation(s)
- Yihao Li
- Gertrude H. Sergievsky Center, Dept of Neurology, Columbia University Vagelos College of Physicians and Surgeons, 630 West 168 Street, PH19-306, New York, N.Y.10032
- Columbia University Vagelos College of Physicians and Surgeons, Department of Medicine, Division of Preventive Medicine and Nutrition, P&S 10-501,New York, NY, USA
| | - Florian Kronenberg
- Institute of Genetic Epidemiology, Medical University of Innsbruck, Innsbruck, Austria
| | - Stefan Coassin
- Institute of Genetic Epidemiology, Medical University of Innsbruck, Innsbruck, Austria
| | - Badri Vardarajan
- Gertrude H. Sergievsky Center, Dept of Neurology, Columbia University Vagelos College of Physicians and Surgeons, 630 West 168 Street, PH19-306, New York, N.Y.10032
| | - Gissette Reyes-Soffer
- Columbia University Vagelos College of Physicians and Surgeons, Department of Medicine, Division of Preventive Medicine and Nutrition, P&S 10-501,New York, NY, USA
| |
Collapse
|
8
|
Delgado-Lista J, Mostaza JM, Arrobas-Velilla T, Blanco-Vaca F, Masana L, Pedro-Botet J, Perez-Martinez P, Civeira F, Cuende-Melero JI, Gomez-Barrado JJ, Lahoz C, Pintó X, Suarez-Tembra M, Lopez-Miranda J, Guijarro C. Consensus on lipoprotein(a) of the Spanish Society of Arteriosclerosis. Literature review and recommendations for clinical practice. CLINICA E INVESTIGACION EN ARTERIOSCLEROSIS : PUBLICACION OFICIAL DE LA SOCIEDAD ESPANOLA DE ARTERIOSCLEROSIS 2024; 36:243-266. [PMID: 38599943 DOI: 10.1016/j.arteri.2024.03.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Accepted: 03/13/2024] [Indexed: 04/12/2024]
Abstract
The irruption of lipoprotein(a) (Lp(a)) in the study of cardiovascular risk factors is perhaps, together with the discovery and use of proprotein convertase subtilisin/kexin type 9 (iPCSK9) inhibitor drugs, the greatest novelty in the field for decades. Lp(a) concentration (especially very high levels) has an undeniable association with certain cardiovascular complications, such as atherosclerotic vascular disease (AVD) and aortic stenosis. However, there are several current limitations to both establishing epidemiological associations and specific pharmacological treatment. Firstly, the measurement of Lp(a) is highly dependent on the test used, mainly because of the characteristics of the molecule. Secondly, Lp(a) concentration is more than 80% genetically determined, so that, unlike other cardiovascular risk factors, it cannot be regulated by lifestyle changes. Finally, although there are many promising clinical trials with specific drugs to reduce Lp(a), currently only iPCSK9 (limited for use because of its cost) significantly reduces Lp(a). However, and in line with other scientific societies, the SEA considers that, with the aim of increasing knowledge about the contribution of Lp(a) to cardiovascular risk, it is relevant to produce a document containing the current status of the subject, recommendations for the control of global cardiovascular risk in people with elevated Lp(a) and recommendations on the therapeutic approach to patients with elevated Lp(a).
Collapse
Affiliation(s)
- Javier Delgado-Lista
- Unidad de Lípidos y Aterosclerosis, Servicio de Medicina Interna, Hospital Universitario Reina Sofía; Departamento de Ciencias Médicas y Quirúrgicas, Universidad de Córdoba; IMIBIC, Córdoba; CIBER Fisiopatología Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, España.
| | - Jose M Mostaza
- Unidad de Lípidos y Riesgo Vascular, Servicio de Medicina Interna, Hospital Universitario La Paz, Madrid, España
| | - Teresa Arrobas-Velilla
- Sociedad Española de Medicina de Laboratorio (SEQCML), Laboratorio de Bioquímica Clínica, Hospital Universitario Virgen Macarena, Sevilla, España
| | - Francisco Blanco-Vaca
- Departamento de Bioquímica Clínica, Hospital de la Santa Creu i Sant Pau, Institut d'Investigació Biomèdica Sant Pau (IIB Sant Pau), Barcelona; Departamento de Bioquímica y Biología Molecular, Universitat Autònoma de Barcelona, 08193 Barcelona; CIBER de Diabetes y Enfermedades Metabólicas Asociadas, Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Madrid, España
| | - Luis Masana
- Unidad de Medicina Vascular y Metabolismo, Hospital Universitari Sant Joan, Universitat Rovira i Virgili, IISPV, CIBERDEM, Reus, Tarragona, España
| | - Juan Pedro-Botet
- Unidad de Lípidos y Riesgo Vascular, Servicio de Endocrinología y Nutrición, Hospital del Mar, Barcelona; Departamento de Medicina, Universidad Autónoma de Barcelona, Barcelona, España
| | - Pablo Perez-Martinez
- Unidad de Lípidos y Aterosclerosis, Servicio de Medicina Interna, Hospital Universitario Reina Sofía; Departamento de Ciencias Médicas y Quirúrgicas, Universidad de Córdoba; IMIBIC, Córdoba; CIBER Fisiopatología Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, España
| | - Fernando Civeira
- Unidad Clínica y de Investigación en Lípidos y Arteriosclerosis, Servicio de Medicina Interna, Hospital Universitario Miguel Servet, IIS Aragón, Universidad de Zaragoza, Zaragoza; CIBER Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, Madrid, España
| | - Jose I Cuende-Melero
- Consulta de Riesgo Vascular, Servicio de Medicina Interna, Complejo Asistencial Universitario de Palencia, Palencia; Departamento de Medicina, Dermatología y Toxicología, Facultad de Medicina, Universidad de Valladolid, Valladolid, España
| | - Jose J Gomez-Barrado
- Unidad de Cuidados Cardiológicos Agudos y Riesgo Cardiovascular, Servicio de Cardiología, Hospital Universitario San Pedro de Alcántara, Cáceres, España
| | - Carlos Lahoz
- Unidad de Lípidos y Arteriosclerosis, Servicio de Medicina Interna, Hospital La Paz-Carlos III, Madrid, España
| | - Xavier Pintó
- Unidad de Lípidos y Riesgo Vascular, Servicio de Medicina Interna, Hospital Universitario de Bellvitge-Idibell-Universidad de Barcelona-CiberObn, España
| | - Manuel Suarez-Tembra
- Unidad de Lípidos y RCV, Servicio de Medicina Interna, Hospital San Rafael, A Coruña, España
| | - Jose Lopez-Miranda
- Unidad de Lípidos y Aterosclerosis, Servicio de Medicina Interna, Hospital Universitario Reina Sofía; Departamento de Ciencias Médicas y Quirúrgicas, Universidad de Córdoba; IMIBIC, Córdoba; CIBER Fisiopatología Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, España.
| | - Carlos Guijarro
- Unidad de Medicina Interna, Hospital Universitario Fundación Alcorcón, Universidad Rey Juan Carlos, Alcorcón, Madrid, España
| |
Collapse
|
9
|
Pablo C, Matías A, Lavalle Cobo A, Sergio G, Federico RN. Exploring the Interplay between Diabetes and Lp(a): Implications for Cardiovascular Risk. Curr Diab Rep 2024; 24:167-172. [PMID: 38805111 DOI: 10.1007/s11892-024-01543-5] [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] [Accepted: 05/15/2024] [Indexed: 05/29/2024]
Abstract
PURPOSE OF REVIEW The objective of this manuscript is to review and describe the relationship between Lp(a) and diabetes, exploring both their association and synergy as cardiovascular risk factors, while also describing the current evidence regarding the potential connection between low levels of Lp(a) and the presence of diabetes. RECENT FINDINGS Epidemiological studies suggest a potential relationship between low to very low levels of Lp(a) and diabetes. Lipoprotein(a), or Lp(a), is an intriguing lipoprotein of genetic origin, yet its biological function remains unknown. Elevated levels of Lp(a) are associated with an increased risk of cardiovascular atherosclerosis, and coexisting diabetes status confers an even higher risk. On the other hand, epidemiological and genetic studies have paradoxically suggested a potential relationship between low to very low levels of Lp(a) and diabetes. While new pharmacological strategies are being developed to reduce Lp(a) levels, the dual aspects of this lipoprotein's behavior need to be elucidated in the near future.
Collapse
Affiliation(s)
- Corral Pablo
- Pharmacology and Research Department, FASTA University, Mar del Plata, Argentina.
| | - Arrupe Matías
- Cardiometabolic Unit Coordinator - Hospital Español, Mendoza, Argentina
| | | | | | - Renna Nicolás Federico
- Chief of Coronary Care Unit - Hospital Español de Mendoza- School of Medicine-UNCuyo, Mendoza, Argentina
| |
Collapse
|
10
|
Liu W, Zhang G, Nie Z, Guan X, Sun T, Jin X, Li B. Low Concentration of Lipoprotein(a) is an Independent Predictor of Incident Type 2 Diabetes. Horm Metab Res 2024; 56:504-508. [PMID: 38772392 DOI: 10.1055/a-2316-9124] [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] [Indexed: 05/23/2024]
Abstract
The aim of the study was to assess the association between lipoprotein(a) [Lp(a)] concentration and incident type 2 diabetes. A meta-analysis of qualified studies on the relationship of low levels of Lp(a) concentration with incident type 2 diabetes was conducted. PubMed and Cochrane libraries were searched for randomized controlled trials containing data on events. Seven randomized trials with 227178 subjects were included in this analysis. We found an inverse association of the levels of Lp(a) concentration with risk of type 2 diabetes with approximately 37% lower relative risk in the group with the highest concentration compared with group with the lowest concentration. The current available evidence from prospective studies suggests that there is an inverse association between the levels of Lp(a) concentration and risk of type 2 diabetes, with a higher risk of type 2 diabetes at low levels of Lp(a) concentration. Therefore, we believe that the low levels of Lp(a) concentration is an independent predictor of incident type 2 diabetes.
Collapse
Affiliation(s)
- Wenhao Liu
- Cardiology, Zibo Central Hospital, Zibo, China
| | | | - Zifan Nie
- Cardiology, Zibo Central Hospital, Zibo, China
| | - Xiangfeng Guan
- Cardiology, Shandong Second Medical University, Weifang, China
| | - Tingting Sun
- Cardiology, Shandong Second Medical University, Weifang, China
| | - Xiaodong Jin
- Geriatric Medicine, Zibo Central Hospital, Zibo, China
| | - Bo Li
- Cardiology, Zibo Central Hospital, Zibo, China
| |
Collapse
|
11
|
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
|
12
|
Béliard S, Mourre F, Valéro R. Hyperlipidaemia in diabetes: are there particular considerations for next-generation therapies? Diabetologia 2024; 67:974-984. [PMID: 38376536 PMCID: PMC11058750 DOI: 10.1007/s00125-024-06100-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/05/2023] [Accepted: 12/07/2023] [Indexed: 02/21/2024]
Abstract
Dyslipidaemias are major cardiovascular risk factors, especially in people with diabetes. In this area, next-generation therapies targeting circulating lipoparticle metabolism (LDL, VLDL, chylomicrons, HDL) have recently been approved by the European and US medical agencies, including anti- proprotein convertase subtilisin/kexin 9 (PCSK9) antibodies; an siRNA targeting PCSK9; bempedoic acid, which targets ATP citrate lyase; an antisense oligonucleotide targeting apolipoprotein C-III; an anti-angiopoietin-like 3 antibody; and a purified omega-3 fatty acid, icosapent ethyl. Other therapies are in different phases of development. There are several important considerations concerning the link between these new lipid-lowering therapies and diabetes. First, since concerns were first raised in 2008 about an increased risk of new-onset diabetes mellitus (NODM) with intensive statin treatment, each new lipid-lowering therapy is being evaluated for its associated risk of NODM, particularly in individuals with prediabetes (impaired fasting glucose and/or impaired glucose tolerance). Second, people with diabetes represent a large proportion of those at high or very high cardiovascular risk in whom these lipid-lowering drugs are currently, or will be, prescribed. Thus, the efficacy of these drugs in subgroups with diabetes should also be closely considered, as well as any potential effects on glycaemic control. In this review, we describe the efficacy of next-generation therapies targeting lipoprotein metabolism in subgroups of people with diabetes and their effects on glycaemic control in individuals with diabetes and prediabetes and in normoglycaemic individuals.
Collapse
Affiliation(s)
- Sophie Béliard
- APHM (Assistance Publique-Hôpitaux de Marseille), Department of Nutrition, Metabolic Diseases, Endocrinology, La Conception Hospital, Marseille, France.
- Inserm, INRAE (Institut National de Recherche pour l'agriculture, l'Alimentation et l'Environnement), C2VN (Centre de recherche en CardioVasculaire et Nutrition), Aix Marseille University, Marseille, France.
| | - Florian Mourre
- APHM (Assistance Publique-Hôpitaux de Marseille), Department of Nutrition, Metabolic Diseases, Endocrinology, La Conception Hospital, Marseille, France
- Inserm, INRAE (Institut National de Recherche pour l'agriculture, l'Alimentation et l'Environnement), C2VN (Centre de recherche en CardioVasculaire et Nutrition), Aix Marseille University, Marseille, France
| | - René Valéro
- APHM (Assistance Publique-Hôpitaux de Marseille), Department of Nutrition, Metabolic Diseases, Endocrinology, La Conception Hospital, Marseille, France
- Inserm, INRAE (Institut National de Recherche pour l'agriculture, l'Alimentation et l'Environnement), C2VN (Centre de recherche en CardioVasculaire et Nutrition), Aix Marseille University, Marseille, France
| |
Collapse
|
13
|
Thazhe Poyil NJ, Vadakkekuttical RJ, Radhakrishnan C. Correlation of periodontal inflamed surface area with glycated hemoglobin, interleukin-6 and lipoprotein(a) in type 2 diabetes with retinopathy. World J Diabetes 2024; 15:686-696. [PMID: 38680698 PMCID: PMC11045419 DOI: 10.4239/wjd.v15.i4.686] [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: 12/20/2023] [Revised: 01/19/2024] [Accepted: 03/01/2024] [Indexed: 04/11/2024] Open
Abstract
BACKGROUND The two-way relationship between periodontitis and type 2 diabetes mellitus (T2DM) is well established. Prolonged hyperglycemia contributes to increased periodontal destruction and severe periodontitis, accentuating diabetic complications. An inflammatory link exists between diabetic retinopathy (DR) and periodontitis, but the studies regarding this association and the role of lipoprotein(a) [Lp(a)] and interleukin-6 (IL-6) in these conditions are scarce in the literature. AIM To determine the correlation of periodontal inflamed surface area (PISA) with glycated Hb (HbA1c), serum IL-6 and Lp(a) in T2DM subjects with retinopathy. METHODS This cross-sectional study comprised 40 T2DM subjects with DR and 40 T2DM subjects without DR. All subjects were assessed for periodontal parameters [bleeding on probing (BOP), probing pocket depth, clinical attachment loss (CAL), oral hygiene index-simplified, plaque index (PI) and PISA], and systemic parameters [HbA1c, fasting plasma glucose and postprandial plasma glucose, fasting lipid profile, serum IL-6 and serum Lp(a)]. RESULTS The proportion of periodontitis in T2DM with and without DR was 47.5% and 27.5% respectively. Severity of periodontitis, CAL, PISA, IL-6 and Lp(a) were higher in T2DM with DR group compared to T2DM without DR group. Sig-nificant difference was observed in the mean percentage of sites with BOP between T2DM with DR (69%) and T2DM without DR (41%), but there was no significant difference in PI (P > 0.05). HbA1c was positively correlated with CAL (r = 0.351, P = 0.001), and PISA (r = 0.393, P ≤ 0.001) in study subjects. A positive correlation was found between PISA and IL-6 (r = 0.651, P < 0.0001); PISA and Lp(a) (r = 0.59, P < 0.001); CAL and IL-6 (r = 0.527, P < 0.0001) and CAL and Lp(a) (r = 0.631, P < 0.001) among study subjects. CONCLUSION Despite both groups having poor glycemic control and comparable plaque scores, the periodontal parameters were higher in DR as compared to T2DM without DR. Since a bidirectional link exists between periodontitis and DM, the presence of DR may have contributed to the severity of periodontal destruction and periodontitis may have influenced the progression of DR.
Collapse
Affiliation(s)
- Nusreen Jamal Thazhe Poyil
- Department of Periodontics, Government Dental College (Affiliated to Kerala University of Health Sciences), Calicut 673008, Kerala, India
| | - Rosamma Joseph Vadakkekuttical
- Department of Periodontics, Government Dental College (Affiliated to Kerala University of Health Sciences), Calicut 673008, Kerala, India
| | - Chandni Radhakrishnan
- Department of Medicine, Government Medical College (Affiliated to Kerala University of Health Sciences), Calicut 673008, Kerala, India
| |
Collapse
|
14
|
Laffin LJ, Nissen SE. Lp(a) - an overlooked risk factor. Trends Cardiovasc Med 2024; 34:193-199. [PMID: 36681362 DOI: 10.1016/j.tcm.2023.01.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Revised: 12/09/2022] [Accepted: 01/14/2023] [Indexed: 01/20/2023]
Abstract
Lipoprotein(a) (Lp(a)) is an increasingly discussed and studied risk factor for atherosclerotic cardiovascular disease and aortic valve stenosis. Many genetic and epidemiological studies support the important causal role that Lp(a) plays in the incidence of cardiovascular disease. Although dependent upon the threshold and unit of measurement of Lp(a), most estimates suggest between 20 and 30% of the world's population have elevated serum levels of Lp(a). Lp(a) levels are predominantly mediated by genetics and are not significantly modified by lifestyle interventions. Efforts are ongoing to develop effective pharmacotherapies to lower Lp(a) and to determine if lowering Lp(a) with these medications ultimately decreases the incidence of adverse cardiovascular events. In this review, the genetics and pathophysiological properties of Lp(a) will be discussed as well as the epidemiological data demonstrating its impact on the incidence of cardiovascular disease. Recommendations for screening and how to currently approach patients with elevated Lp(a) are also noted. Finally, the spectrum of pharmacotherapies under development for Lp(a) lowering is detailed.
Collapse
|
15
|
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
|
16
|
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
|
17
|
Chiesa G, Zenti MG, Baragetti A, Barbagallo CM, Borghi C, Colivicchi F, Maggioni AP, Noto D, Pirro M, Rivellese AA, Sampietro T, Sbrana F, Arca M, Averna M, Catapano AL. Consensus document on Lipoprotein(a) from the Italian Society for the Study of Atherosclerosis (SISA). Nutr Metab Cardiovasc Dis 2023; 33:1866-1877. [PMID: 37586921 DOI: 10.1016/j.numecd.2023.07.019] [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: 06/14/2023] [Revised: 07/07/2023] [Accepted: 07/13/2023] [Indexed: 08/18/2023]
Abstract
AIMS In view of the consolidating evidence on the causal role of Lp(a) in cardiovascular disease, the Italian Society for the Study of Atherosclerosis (SISA) has assembled a consensus on Lp(a) genetics and epidemiology, together with recommendations for its measurement and current and emerging therapeutic approaches to reduce its plasma levels. Data on the Italian population are also provided. DATA SYNTHESIS Lp(a) is constituted by one apo(a) molecule and a lipoprotein closely resembling to a low-density lipoprotein (LDL). Its similarity with an LDL, together with its ability to carry oxidized phospholipids are considered the two main features making Lp(a) harmful for cardiovascular health. Plasma Lp(a) concentrations vary over about 1000 folds in humans and are genetically determined, thus they are quite stable in any individual. Mendelian Randomization studies have suggested a causal role of Lp(a) in atherosclerotic cardiovascular disease (ASCVD) and aortic valve stenosis and observational studies indicate a linear direct correlation between cardiovascular disease and Lp(a) plasma levels. Lp(a) measurement is strongly recommended once in a patient's lifetime, particularly in FH subjects, but also as part of the initial lipid screening to assess cardiovascular risk. The apo(a) size polymorphism represents a challenge for Lp(a) measurement in plasma, but new strategies are overcoming these difficulties. A reduction of Lp(a) levels can be currently attained only by plasma apheresis and, moderately, with PCSK9 inhibitor treatment. CONCLUSIONS Awaiting the approval of selective Lp(a)-lowering drugs, an intensive management of the other risk factors for individuals with elevated Lp(a) levels is strongly recommended.
Collapse
Affiliation(s)
- Giulia Chiesa
- Department of Pharmacological and Biomolecular Sciences "Rodolfo Paoletti", Università Degli Studi di Milano, Milan, Italy.
| | - Maria Grazia Zenti
- Section of Diabetes and Metabolism, Pederzoli Hospital, Peschiera Del Garda, Verona, Italy.
| | - Andrea Baragetti
- Department of Pharmacological and Biomolecular Sciences "Rodolfo Paoletti", Università Degli Studi di Milano, Milan, Italy; IRCCS MultiMedica, Sesto San Giovanni, Milan, Italy
| | - Carlo M Barbagallo
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties (PROMISE), University of Palermo, Palermo, Italy
| | - Claudio Borghi
- Department of Cardiovascular Medicine, IRCCS AOU S. Orsola, Bologna, Italy
| | - Furio Colivicchi
- Division of Clinical Cardiology, San Filippo Neri Hospital, Rome, Italy
| | - Aldo P Maggioni
- ANMCO Research Center, Heart Care Foundation, Firenze, Italy
| | - Davide Noto
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties (PROMISE), University of Palermo, Palermo, Italy
| | - Matteo Pirro
- Unit of Internal Medicine, Angiology and Arteriosclerosis Diseases, Department of Medicine and Surgery, University of Perugia, Italy
| | - Angela A Rivellese
- Department of Clinical Medicine and Surgery, Federico II University, Naples, Italy
| | - Tiziana Sampietro
- Lipoapheresis Unit, Reference Center for Diagnosis and Treatment of Inherited Dyslipidemias, Fondazione Toscana Gabriele Monasterio, Pisa, Italy
| | - Francesco Sbrana
- Lipoapheresis Unit, Reference Center for Diagnosis and Treatment of Inherited Dyslipidemias, Fondazione Toscana Gabriele Monasterio, Pisa, Italy
| | - Marcello Arca
- Department of Translational and Precision Medicine (DTPM), Sapienza University of Rome, Policlinico Umberto I, Rome, Italy
| | - Maurizio Averna
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties (PROMISE), University of Palermo, Palermo, Italy; Institute of Biophysics, National Council of Researches, Palermo, Italy
| | - Alberico L Catapano
- Department of Pharmacological and Biomolecular Sciences "Rodolfo Paoletti", Università Degli Studi di Milano, Milan, Italy; IRCCS MultiMedica, Sesto San Giovanni, Milan, Italy
| |
Collapse
|
18
|
Wang Y, Bulloch G, Huang Y, Liang Y, Du Z, Wu G, Fang Y, Hu Y, Shang X, Zhu Z, Zhang X, Yang X, Yu H. Determinants of incident atherosclerotic cardiovascular disease events among individuals with type 2 diabetic microvascular complications in the UK: a prospective cohort study. Diabetol Metab Syndr 2023; 15:178. [PMID: 37644580 PMCID: PMC10463420 DOI: 10.1186/s13098-023-01152-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Accepted: 08/19/2023] [Indexed: 08/31/2023] Open
Abstract
OBJECTIVE To evaluate the association of atherosclerotic cardiovascular disease (ASCVD) risk factors with incident ASCVD events among type 2 diabetes (T2D) individuals with microvascular complications. METHODS We included T2D participants with only microvascular complications from the UK Biobank cohort at baseline (2006-2010). Multivariable-adjusted Cox proportional hazards models were used to study the association between ASCVD risk factors with adjudicated incident ASCVD in T2D participants with only microvascular complications. A restricted cubic spline approach was employed to evaluate potential nonlinear associations between ASCVD risk factors and ASCVD. RESULTS We studied 4,129 T2D individuals with microvascular complications at baseline. Over a median follow-up of 11.7 years, a total of 1,180 cases of incident ASCVD were documented, of which 1,040 were CHD, 100 were stroke, and 40 were both CHD and stroke events. After multivariable-adjustment, high-density lipoprotein cholesterol (HDL-C) level was linearly associated with a decreased risk of incident ASCVD [hazard ratio (HR): 0.49, 95% Confidence interval (CI): 0.32-0.75, Plinear = 0.011] and each 10 nmol/L increase of lipoprotein(a) [Lp(a)] level (HR: 1.02, 95% CI: 1.00-1.04, Plinear = 0.012) was linearly associated with an increased risk of incident ASCVD in T2D participants with only microvascular complications. CONCLUSION HDL-C levels and Lp(a) levels (per 10 nmol/L) showed an independent linear relation with ASCVD risk among T2D individuals with only microvascular complications at long-term follow-up.
Collapse
Affiliation(s)
- Yaxin Wang
- Guangdong Eye Institute, Department of Ophthalmology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Gabriella Bulloch
- Centre for Eye Research Australia, Royal Victorian Eye and Ear Hospital, East Melbourne, VIC, Australia
| | - Yu Huang
- Guangdong Eye Institute, Department of Ophthalmology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Yingying Liang
- Guangdong Eye Institute, Department of Ophthalmology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Zijing Du
- Guangdong Eye Institute, Department of Ophthalmology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Guanrong Wu
- Guangdong Eye Institute, Department of Ophthalmology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Ying Fang
- Guangdong Eye Institute, Department of Ophthalmology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Yijun Hu
- Guangdong Eye Institute, Department of Ophthalmology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Xianwen Shang
- Guangdong Eye Institute, Department of Ophthalmology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
- Centre for Eye Research Australia, Royal Victorian Eye and Ear Hospital, East Melbourne, VIC, Australia
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Zhuoting Zhu
- Guangdong Eye Institute, Department of Ophthalmology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
- Centre for Eye Research Australia, Royal Victorian Eye and Ear Hospital, East Melbourne, VIC, Australia
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Xiayin Zhang
- Guangdong Eye Institute, Department of Ophthalmology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China.
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China.
| | - Xiaohong Yang
- Guangdong Eye Institute, Department of Ophthalmology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China.
| | - Honghua Yu
- Guangdong Eye Institute, Department of Ophthalmology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China.
- Guangdong Provincial Key Laboratory of Artificial Intelligence in Medical Image Analysis and Application, Guangzhou, China.
| |
Collapse
|
19
|
Yu B, Hu X, Liu J, Nie Z, Ren Luo Bu C, Li G, Zhou Y, Dong H. Lipoprotein(a) as a Higher Residual Risk for Coronary Artery Disease in Patients with Type 2 Diabetes Mellitus than without. Int J Gen Med 2023; 16:3383-3391. [PMID: 37576912 PMCID: PMC10422995 DOI: 10.2147/ijgm.s423458] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2023] [Accepted: 07/31/2023] [Indexed: 08/15/2023] Open
Abstract
Purpose Lipoprotein(a) (Lp[a]) is well-known as a residual risk factor for coronary artery disease (CAD). However, the different adverse effects of Lp(a) about CAD in patients with or without type 2 diabetes mellitus (T2DM) are unclear. This study aimed to investigate the Lp(a) thresholds for CAD diagnosis in T2DM and non-T2DM patients, and further compare the Lp(a) alarm values along with optimal low-density lipoprotein cholesterol (LDL-C) level. Methods This retrospective study consecutively enrolled patients with suspected CAD who underwent coronary angiography in Guangdong Provincial People's Hospital between September 2014 and July 2015. A logistic regression model was established to explore the association of Lp(a) and CAD in patients. Restricted cubic splines were used to compare the threshold values of Lp(a) for CAD in patients with and without T2DM, and further in optimal LDL-C level situation. Results There were 1522 patients enrolled finally. After multivariable adjustment, Lp(a) was an independent risk factor for CAD in patients with T2DM (odds ratio [OR]: 1.98, 95% CI]: 1.12-3.49, p = 0.019) and without T2DM (OR: 3.42, 95% CI: 2.36-4.95, p < 0.001). In the whole population, the Lp(a) threshold of CAD was 155, while 145 mg/L for T2DM and 162 mg/L for non-T2DM ones, respectively. In patients with LDL-C<1.8 mmol/l, the alarm value of Lp(a) was even lower in T2DM than non-T2DM patients (155 vs 174 mg/L). Conclusion Lp(a) was a significant residual risk for CAD in patients whether with T2DM or not. And Lp(a) had a lower alarm value in T2DM patients, especially in optimal LDL-C level.
Collapse
Affiliation(s)
- Bingyan Yu
- School of Medicine, South China University of Technology, Guangzhou, People’s Republic of China
- Department of Cardiology, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, People’s Republic of China
| | - Xiangming Hu
- Department of Cardiology, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, People’s Republic of China
| | - Jieliang Liu
- Department of Cardiology, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, People’s Republic of China
| | - Zhiqiang Nie
- Department of Cardiology, Hypertension Research Laboratory, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, People’s Republic of China
| | - Ci Ren Luo Bu
- Nyingchi People’s Hospital, Nyingchi, Tibet, People’s Republic of China
| | - Guang Li
- Department of Cardiology, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, People’s Republic of China
| | - Yingling Zhou
- Department of Cardiology, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, People’s Republic of China
| | - Haojian Dong
- Department of Cardiology, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, People’s Republic of China
- Nyingchi People’s Hospital, Nyingchi, Tibet, People’s Republic of China
| |
Collapse
|
20
|
Skoumas I, Andrikou I, Grigoriou K, Dima I, Lazarou E, Vlachopoulos C, Tsioufis K. Lipoprotein(a), metabolic profile and new-onset type 2 diabetes in patients with familial combined hyperlipidemia: A 9 year follow-up study. J Clin Lipidol 2023; 17:512-518. [PMID: 37321915 DOI: 10.1016/j.jacl.2023.05.103] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Revised: 05/17/2023] [Accepted: 05/29/2023] [Indexed: 06/17/2023]
Abstract
BACKGROUND Lipoprotein(a) [Lp(a)] appears to have an inverse association with the risk of type 2 diabetes mellitus in the general population. OBJECTIVE This study aimed to investigate the prognostic role of Lp(a) regarding the development of type 2 diabetes in the special population of subjects with familial combined hyperlipidemia (FCH). METHODS This cohort study included 474 patients (mean age 49.7±11.3 years, 64% males) with FCH, without diabetes at baseline who were followed for a mean period of 8.2±6.8 years. At baseline evaluation venous blood samples were obtained for the determination of lipid profile and Lp(a) levels. The endpoint of interest was the development of diabetes. RESULTS Patients with increased Lp(a) levels ≥30 mg/dl compared to those with low Lp(a) levels <30 mg/dl had lower levels of triglycerides (238±113 vs 268±129 mg/dl, p = 0.01), greater levels of high-density lipoprotein (HDL) cholesterol (44±10 vs 41±10 mg/dl, p = 0.01) and hypertension in a greater percentage (42% vs 32%, p = 0.03). The incidence of new-onset diabetes during the follow-up period was 10.1% (n = 48). Multiple Cox regression analysis revealed that increased Lp(a) is an independent predictor of lower diabetes incidence (HR 0.39, 95% CI 0.17-0.90, p = 0.02) after adjustment for confounders. CONCLUSION Among subjects with FCH those with higher Lp(a) levels have lower risk for the development of type 2 diabetes. Moreover, the presence of increased Lp(a) seems to differentiate the expression of metabolic syndrome characteristics in patients with FCH, as increased Lp(a) is related to lower levels of triglycerides, greater prevalence of hypertension and higher levels of HDL cholesterol.
Collapse
Affiliation(s)
- Ioannis Skoumas
- First Department of Cardiology, Hippokration Hospital, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Ioannis Andrikou
- First Department of Cardiology, Hippokration Hospital, Medical School, National and Kapodistrian University of Athens, Athens, Greece.
| | - Kalliopi Grigoriou
- First Department of Cardiology, Hippokration Hospital, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Ioanna Dima
- First Department of Cardiology, Hippokration Hospital, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Emilia Lazarou
- First Department of Cardiology, Hippokration Hospital, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Charalambos Vlachopoulos
- First Department of Cardiology, Hippokration Hospital, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Konstantinos Tsioufis
- First Department of Cardiology, Hippokration Hospital, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| |
Collapse
|
21
|
Chen F, Fei X, Zhu W, Zhang Z, Shen Y, Mao Y, Zhu Q, Xu J, Zhou W, Li M, Du J. Placental DNA methylation changes in gestational diabetes mellitus. Epigenetics 2022; 17:2109-2121. [PMID: 35993280 PMCID: PMC9665131 DOI: 10.1080/15592294.2022.2110193] [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: 03/26/2022] [Accepted: 08/01/2022] [Indexed: 11/03/2022] Open
Abstract
In this study, we investigated the association between altered methylation in the maternal placenta and hyperglycaemia and explored the epigenetic mechanisms underlying gestational diabetes mellitus (GDM). Reduced representation bisulphite sequencing (RRBS) and RNA sequencing (RNA-seq) were performed on placental tissues obtained from women with GDM and healthy controls. Further, pyrosequencing, correlation analyses, and linear regression analyses were performed to valuate relationships between aberrantly methylated-differentially expressed genes and clinical parameters. The EMBOSS and JASPAR databases were used for a computational analysis of CpG islands and transcription factor-binding sites in the TRIM67 promoter region. A CpG island with a length of 264 bp in the placental TRIM67 promoter region in the GDM group exhibited significant hypermethylation at four CpG sites. The hypermethylation of the TRIM67 promoter region in the maternal placenta showed a significant, positive correlation with the 1 h and 2 h oral glucose tolerance test (OGTT) values and a negative correlation with lipoprotein(a). Placental DNA methylation levels in the TRIM67 promoter region were markedly elevated in GDM and were associated with blood glucose and lipid levels during healthy pregnancy.
Collapse
Affiliation(s)
- Fujia Chen
- NHC Key Lab of Reproduction Regulation (Shanghai Institute for Biomedical and Pharmaceutical Technologies), School of Pharmacy, Fudan University, Shanghai, China
| | - Xiaoping Fei
- Obstetrics Department, The First People’s Hospital of Kunshan, Kunshan, China
| | - Weiqiang Zhu
- NHC Key Lab of Reproduction Regulation (Shanghai Institute for Biomedical and Pharmaceutical Technologies), School of Pharmacy, Fudan University, Shanghai, China
| | - Zhaofeng Zhang
- NHC Key Lab of Reproduction Regulation (Shanghai Institute for Biomedical and Pharmaceutical Technologies), School of Pharmacy, Fudan University, Shanghai, China
| | - Yupei Shen
- NHC Key Lab of Reproduction Regulation (Shanghai Institute for Biomedical and Pharmaceutical Technologies), School of Pharmacy, Fudan University, Shanghai, China
| | - Yanyan Mao
- NHC Key Lab of Reproduction Regulation (Shanghai Institute for Biomedical and Pharmaceutical Technologies), School of Pharmacy, Fudan University, Shanghai, China
| | - Qianxi Zhu
- NHC Key Lab of Reproduction Regulation (Shanghai Institute for Biomedical and Pharmaceutical Technologies), School of Pharmacy, Fudan University, Shanghai, China
| | - Jianhua Xu
- NHC Key Lab of Reproduction Regulation (Shanghai Institute for Biomedical and Pharmaceutical Technologies), School of Pharmacy, Fudan University, Shanghai, China
| | - Weijin Zhou
- NHC Key Lab of Reproduction Regulation (Shanghai Institute for Biomedical and Pharmaceutical Technologies), School of Pharmacy, Fudan University, Shanghai, China
| | - Min Li
- NHC Key Lab of Reproduction Regulation (Shanghai Institute for Biomedical and Pharmaceutical Technologies), School of Pharmacy, Fudan University, Shanghai, China
| | - Jing Du
- NHC Key Lab of Reproduction Regulation (Shanghai Institute for Biomedical and Pharmaceutical Technologies), School of Pharmacy, Fudan University, Shanghai, China
| |
Collapse
|
22
|
Nestel P, Loh WJ, Ward NC, Watts GF. New Horizons: Revival of Lipoprotein (a) as a Risk Factor for Cardiovascular Disease. J Clin Endocrinol Metab 2022; 107:e4281-e4294. [PMID: 36108076 DOI: 10.1210/clinem/dgac541] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/09/2022] [Indexed: 02/13/2023]
Abstract
The status of lipoprotein (a) [Lp(a)] as a cardiovascular risk factor has been resurrected by advances in genetics. Mendelian randomization studies show a causal link of Lp(a) with coronary artery disease (CAD), peripheral artery disease (PAD), and calcific aortic valve stenosis (CAVS). The genetics of Lp(a) is complex and extends beyond the kringle-IV type 2, as it is also dependent on ancestry. The plasma concentration of Lp(a) is determined by the hepatic production of apolipoprotein(a) [apo(a)] component of Lp(a), supporting the use of nucleic acids that inhibit the messenger RNA (mRNA) gene transcript for apo(a). Analytical barriers to measurement of Lp(a) are being addressed using isoform independent assays and a traceable standard. The association of Lp(a) and atherosclerotic cardiovascular disease is higher for myocardial infarction than PAD and CAVS. Increased risk of type 2 diabetes mellitus associated with low Lp(a) levels is perplexing and requires further investigation. The greatest advancement in Lp(a)-lowering therapies is based on using RNA therapeutics that are now being investigated in clinical trials. Proprotein convertase subtilisin/kexin type 9 (PCSK9) inhibition lowers Lp(a) modestly, but whether cardiovascular benefit is independent of low-density lipoprotein lowering remains unclear. Opportunistic and selective testing for Lp(a) is supported by moderate evidence, with the case for universal screening premature. Modification of behavioral and clinical risk factors may be targeted to mitigate Lp(a)-mediated risk of cardiovascular disease. Clinical practice guidelines have been developed to address gaps in care of high Lp(a), but full implementation awaits the findings of clinical outcome trials using RNA-directed therapies currently underway.
Collapse
Affiliation(s)
- Paul Nestel
- Baker Heart and Diabetes Institute, Melbourne, Australia
| | - Wann Jia Loh
- School of Medicine, University of Western Australia, Perth, Australia
- Department of Cardiology and Internal Medicine, Royal Perth Hospital, Perth, Australia
- Department of Endocrinology, Changi General Hospital, Singapore
- Duke-NUS Medical School, Singapore
| | - Natalie C Ward
- School of Medicine, University of Western Australia, Perth, Australia
| | - Gerald F Watts
- School of Medicine, University of Western Australia, Perth, Australia
- Department of Cardiology and Internal Medicine, Royal Perth Hospital, Perth, Australia
| |
Collapse
|
23
|
Kronenberg F, Mora S, Stroes ESG, Ference BA, Arsenault BJ, Berglund L, Dweck MR, Koschinsky M, Lambert G, Mach F, McNeal CJ, Moriarty PM, Natarajan P, Nordestgaard BG, Parhofer KG, Virani SS, von Eckardstein A, Watts GF, Stock JK, Ray KK, Tokgözoğlu LS, Catapano AL. Lipoprotein(a) in atherosclerotic cardiovascular disease and aortic stenosis: a European Atherosclerosis Society consensus statement. Eur Heart J 2022; 43:3925-3946. [PMID: 36036785 PMCID: PMC9639807 DOI: 10.1093/eurheartj/ehac361] [Citation(s) in RCA: 394] [Impact Index Per Article: 197.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Revised: 05/10/2022] [Accepted: 06/21/2022] [Indexed: 12/20/2022] Open
Abstract
This 2022 European Atherosclerosis Society lipoprotein(a) [Lp(a)] consensus statement updates evidence for the role of Lp(a) in atherosclerotic cardiovascular disease (ASCVD) and aortic valve stenosis, provides clinical guidance for testing and treating elevated Lp(a) levels, and considers its inclusion in global risk estimation. Epidemiologic and genetic studies involving hundreds of thousands of individuals strongly support a causal and continuous association between Lp(a) concentration and cardiovascular outcomes in different ethnicities; elevated Lp(a) is a risk factor even at very low levels of low-density lipoprotein cholesterol. High Lp(a) is associated with both microcalcification and macrocalcification of the aortic valve. Current findings do not support Lp(a) as a risk factor for venous thrombotic events and impaired fibrinolysis. Very low Lp(a) levels may associate with increased risk of diabetes mellitus meriting further study. Lp(a) has pro-inflammatory and pro-atherosclerotic properties, which may partly relate to the oxidized phospholipids carried by Lp(a). This panel recommends testing Lp(a) concentration at least once in adults; cascade testing has potential value in familial hypercholesterolaemia, or with family or personal history of (very) high Lp(a) or premature ASCVD. Without specific Lp(a)-lowering therapies, early intensive risk factor management is recommended, targeted according to global cardiovascular risk and Lp(a) level. Lipoprotein apheresis is an option for very high Lp(a) with progressive cardiovascular disease despite optimal management of risk factors. In conclusion, this statement reinforces evidence for Lp(a) as a causal risk factor for cardiovascular outcomes. Trials of specific Lp(a)-lowering treatments are critical to confirm clinical benefit for cardiovascular disease and aortic valve stenosis.
Collapse
Affiliation(s)
- Florian Kronenberg
- Institute of Genetic Epidemiology, Medical University of Innsbruck, Innsbruck, Austria
| | - Samia Mora
- Center for Lipid Metabolomics, Division of Preventive Medicine, and Division of Cardiovascular Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Erik S G Stroes
- Department of Vascular Medicine, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
| | - Brian A Ference
- Centre for Naturally Randomized Trials, University of Cambridge, Cambridge, UK
| | - Benoit J Arsenault
- Centre de Recherche de l'Institut Universitaire de Cardiologie et de Pneumologie de Québec, and Department of Medicine, Faculty of Medicine, Université Laval, Québec City, QC, Canada
| | - Lars Berglund
- Department of Internal Medicine, School of Medicine, University of California-Davis, Davis, Sacramento, CA, USA
| | - Marc R Dweck
- British Heart Foundation Centre for Cardiovascular Science, Edinburgh Heart Centre, University of Edinburgh, Chancellors Building, Little France Crescent, Edinburgh EH16 4SB, UK
| | - Marlys Koschinsky
- Robarts Research Institute, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada
| | - Gilles Lambert
- Inserm, UMR 1188 Diabète Athérothrombose Thérapies Réunion Océan Indien (DéTROI), Université de La Réunion, 97400 Saint-Denis de La Reunion, France
| | - François Mach
- Department of Cardiology, Geneva University Hospital, Geneva, Switzerland
| | - Catherine J McNeal
- Division of Cardiology, Department of Internal Medicine, Baylor Scott & White Health, 2301 S. 31st St., USA
| | | | - Pradeep Natarajan
- Cardiovascular Research Center, Massachusetts General Hospital, Harvard Medical School, Boston, and Program in Medical and Population Genetics and Cardiovascular Disease Initiative, Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Børge G Nordestgaard
- Department of Clinical Biochemistry and the Copenhagen General Population Study, Herlev and Gentofte Hospital, Copenhagen University Hospital, Herlev, Denmark.,Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Klaus G Parhofer
- Medizinische Klinik und Poliklinik IV, Ludwigs- Maximilians University Klinikum, Munich, Germany
| | - Salim S Virani
- Section of Cardiovascular Research, Baylor College of Medicine & Michael E. DeBakey Veterans Affairs Medical Center, Houston, TX, USA
| | - Arnold von Eckardstein
- Institute of Clinical Chemistry, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Gerald F Watts
- Medical School, University of Western Australia, and Department of Cardiology, Lipid Disorders Clinic, Royal Perth Hospital, Perth, Australia
| | - Jane K Stock
- European Atherosclerosis Society, Mässans Gata 10, SE-412 51 Gothenburg, Sweden
| | - Kausik K Ray
- Imperial Centre for Cardiovascular Disease Prevention, Department of Primary Care and Public Health, School of Public Health, Imperial College London, London, UK
| | - Lale S Tokgözoğlu
- Department of Cardiology, Hacettepe University Faculty of Medicine, Ankara, Turkey
| | - Alberico L Catapano
- Department of Pharmacological and Biomolecular Sciences, University of Milano, Milano, Italy.,IRCCS Multimedica, Milano, Italy
| |
Collapse
|
24
|
Ramos-Cáceres M, Lamiquiz-Moneo I, Cenarro A, Calmarza P, Marco-Benedí V, Bea AM, Mateo-Gallego R, Puzo J, Ordovas JM, Civeira F, Laclaustra M. Triglyceride Metabolism Modifies Lipoprotein(a) Plasma Concentration. J Clin Endocrinol Metab 2022; 107:e3594-e3602. [PMID: 35789387 DOI: 10.1210/clinem/dgac412] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/24/2022] [Indexed: 11/19/2022]
Abstract
BACKGROUND Lipoprotein(a) (Lp(a)) is a significant cardiovascular risk factor. Knowing the mechanisms that regulate its concentration can facilitate the development of Lp(a)-lowering drugs. This study analyzes the relationship between triglycerides (TGs) and Lp(a) concentrations, cross-sectionally and longitudinally, and the influence of the number and composition of TG-rich lipoproteins, and the APOE genotype. METHODS Data from Aragon Workers Health Study (AWHS) (n = 5467), National Health and Nutrition Examination Survey III phase 2 (n = 3860), and Hospital Universitario Miguel Servet (HUMS) (n = 2079) were used for cross-sectional TG and Lp(a) relationship. Lp(a) intrasubject variation was studied in AWHS participants and HUMS patients with repeated measurements. TG-rich lipoproteins were quantified by nuclear magnetic resonance in a subsample from AWHS. Apolipoproteins B and E were quantified by Luminex in very low-density lipoprotein (VLDL) isolated by ultracentrifugation, from HUMS samples. APOE genotyping was carried in AWHS and HUMS participants. Regression models adjusted for age and sex were used to study the association. RESULTS The 3 studies showed an inverse relationship between TG and Lp(a). Increased VLDL number, size, and TG content were associated with significantly lower Lp(a). There was an inverse association between the apoE concentration in VLDL and Lp(a). No significant association was observed for apolipoprotein (apo)B. Subjects carrying the apoE2/E2 genotype had significantly lower levels of Lp(a). CONCLUSION Our results show an inverse relationship Lp(a)-TG. Subjects with larger VLDL size have lower Lp(a), and lower values of Lp(a) were present in patients with apoE-rich VLDL and apoE2/E2 subjects. Our results suggest that bigger VLDLs and VLDLs enriched in apoE are inversely involved in Lp(a) plasma concentration.
Collapse
Affiliation(s)
- Maria Ramos-Cáceres
- Hospital Universitario Miguel Servet, Instituto de Investigación Sanitaria Aragón (IIS Aragón), CIBERCV, Zaragoza 50009, Spain
| | - Itziar Lamiquiz-Moneo
- Hospital Universitario Miguel Servet, Instituto de Investigación Sanitaria Aragón (IIS Aragón), CIBERCV, Zaragoza 50009, Spain
- Departamento de Anatomía e Histología Humanas, Facultad de Medicina, Universidad de Zaragoza, Zaragoza 50009, Spain
| | - Ana Cenarro
- Hospital Universitario Miguel Servet, Instituto de Investigación Sanitaria Aragón (IIS Aragón), CIBERCV, Zaragoza 50009, Spain
- Instituto Aragonés de Ciencias de la Salud, (IACS), Zaragoza 50009, Spain
| | - Pilar Calmarza
- Hospital Universitario Miguel Servet, Instituto de Investigación Sanitaria Aragón (IIS Aragón), CIBERCV, Zaragoza 50009, Spain
| | - Victoria Marco-Benedí
- Hospital Universitario Miguel Servet, Instituto de Investigación Sanitaria Aragón (IIS Aragón), CIBERCV, Zaragoza 50009, Spain
- Departamento de Medicina, Psiquiatría y Dermatología, Facultad de Medicina, Universidad de Zaragoza, Zaragoza 50009, Spain
| | - Ana M Bea
- Hospital Universitario Miguel Servet, Instituto de Investigación Sanitaria Aragón (IIS Aragón), CIBERCV, Zaragoza 50009, Spain
| | - Rocio Mateo-Gallego
- Hospital Universitario Miguel Servet, Instituto de Investigación Sanitaria Aragón (IIS Aragón), CIBERCV, Zaragoza 50009, Spain
- Departamento de Fisiatría y Enfermería, Facultad de Ciencias de la Salud y del Deporte, Universidad de Zaragoza, Huesca 22002, Spain
| | - Jose Puzo
- Departamento de Medicina, Psiquiatría y Dermatología, Facultad de Medicina, Universidad de Zaragoza, Zaragoza 50009, Spain
- Unidad de Lípidos, Servicio de Análisis y Bioquímica Clínica, Hospital San Jorge, Huesca 22004, Spain
| | - Jose M Ordovas
- Nutrition and Genomics Laboratory, JM-USDA Human Nutrition Research Center on Aging at Tufts University, Boston, Massachusetts 02111, USA
- Precision Nutrition and Obesity Program, IMDEA Alimentación, Madrid 28049, Spain
| | - Fernando Civeira
- Hospital Universitario Miguel Servet, Instituto de Investigación Sanitaria Aragón (IIS Aragón), CIBERCV, Zaragoza 50009, Spain
- Departamento de Medicina, Psiquiatría y Dermatología, Facultad de Medicina, Universidad de Zaragoza, Zaragoza 50009, Spain
| | - Martin Laclaustra
- Hospital Universitario Miguel Servet, Instituto de Investigación Sanitaria Aragón (IIS Aragón), CIBERCV, Zaragoza 50009, Spain
- Departamento de Medicina, Psiquiatría y Dermatología, Facultad de Medicina, Universidad de Zaragoza, Zaragoza 50009, Spain
| |
Collapse
|
25
|
Šuran D, Blažun Vošner H, Završnik J, Kokol P, Sinkovič A, Kanič V, Kokol M, Naji F, Završnik T. Lipoprotein(a) in Cardiovascular Diseases: Insight From a Bibliometric Study. Front Public Health 2022; 10:923797. [PMID: 35865239 PMCID: PMC9294325 DOI: 10.3389/fpubh.2022.923797] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Accepted: 06/07/2022] [Indexed: 11/13/2022] Open
Abstract
Lipoprotein(a) [Lp(a)] is a complex polymorphic lipoprotein comprised of a low-density lipoprotein particle with one molecule of apolipoprotein B100 and an additional apolipoprotein(a) connected through a disulfide bond. The serum concentration is mostly genetically determined and only modestly influenced by diet and other lifestyle modifications. In recent years it has garnered increasing attention due to its causal role in pre-mature atherosclerotic cardiovascular disease and calcific aortic valve stenosis, while novel effective therapeutic options are emerging [apolipoprotein(a) antisense oligonucleotides and ribonucleic acid interference therapy]. Bibliometric descriptive analysis and mapping of the research literature were made using Scopus built-in services. We focused on the distribution of documents, literature production dynamics, most prolific source titles, institutions, and countries. Additionally, we identified historical and influential papers using Reference Publication Year Spectrography (RPYS) and the CRExplorer software. An analysis of author keywords showed that Lp(a) was most intensively studied regarding inflammation, atherosclerosis, cardiovascular risk assessment, treatment options, and hormonal changes in post-menopausal women. The results provide a comprehensive view of the current Lp(a)-related literature with a specific interest in its role in calcific aortic valve stenosis and potential emerging pharmacological interventions. It will help the reader understand broader aspects of Lp(a) research and its translation into clinical practice.
Collapse
Affiliation(s)
- David Šuran
- Department of Cardiology and Angiology, University Medical Centre Maribor, Maribor, Slovenia
- Faculty of Medicine, University of Maribor, Maribor, Slovenia
- *Correspondence: David Šuran
| | - Helena Blažun Vošner
- Community Healthcare Centre Dr. Adolf Drolc Maribor, Maribor, Slovenia
- Faculty of Health and Social Sciences Slovenj Gradec, Slovenj Gradec, Slovenia
- Alma Mater Europaea, Maribor, Slovenia
| | - Jernej Završnik
- Community Healthcare Centre Dr. Adolf Drolc Maribor, Maribor, Slovenia
- Alma Mater Europaea, Maribor, Slovenia
| | - Peter Kokol
- Faculty of Medicine, University of Maribor, Maribor, Slovenia
- Faculty of Electrical Engineering and Computer Science, University of Maribor, Maribor, Slovenia
| | - Andreja Sinkovič
- Faculty of Medicine, University of Maribor, Maribor, Slovenia
- Department of Medical Intensive Care, University Medical Centre Maribor, Maribor, Slovenia
| | - Vojko Kanič
- Department of Cardiology and Angiology, University Medical Centre Maribor, Maribor, Slovenia
- Faculty of Medicine, University of Maribor, Maribor, Slovenia
| | - Marko Kokol
- Faculty of Electrical Engineering and Computer Science, University of Maribor, Maribor, Slovenia
- Semantika Research, Semantika d.o.o., Maribor, Slovenia
| | - Franjo Naji
- Department of Cardiology and Angiology, University Medical Centre Maribor, Maribor, Slovenia
- Faculty of Medicine, University of Maribor, Maribor, Slovenia
| | - Tadej Završnik
- Department of Cardiology and Angiology, University Medical Centre Maribor, Maribor, Slovenia
- Faculty of Medicine, University of Maribor, Maribor, Slovenia
| |
Collapse
|
26
|
Raitakari O, Kivelä A, Pahkala K, Rovio S, Mykkänen J, Ahola-Olli A, Loo BM, Lyytikäinen LP, Lehtimäki T, Kähönen M, Juonala M, Rönnemaa T, Lamina C, Kronenberg F, Viikari J. Long-term tracking and population characteristics of lipoprotein (a) in the cardiovascular risk in young finns study. Atherosclerosis 2022; 356:18-27. [DOI: 10.1016/j.atherosclerosis.2022.07.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Revised: 06/01/2022] [Accepted: 07/13/2022] [Indexed: 11/16/2022]
|
27
|
Chang X, Bian N, Ding X, Li J, An Y, Wang J, Liu J, Wang G. Study on the relationship between hormone and Lp(a) in Chinese overweight/obese patients. BMC Endocr Disord 2022; 22:131. [PMID: 35578222 PMCID: PMC9109374 DOI: 10.1186/s12902-022-01021-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Accepted: 04/06/2022] [Indexed: 11/15/2022] Open
Abstract
BACKGROUND Obesity is a risk factor for metabolic diseases and often influences hormone change. Lipoprotein (a) (Lp(a)) is associated with various metabolic diseases, but there are few studies on the relationship between Lp(a) and hormones in obese patients. This study investigated the the relationship between Lp(a) and hormones in Chinese overweight/obese people. METHODS A total of 410 overweight/obese patients (Body mass index (BMI) ≥ 25 kg/m2) were included and underwent sociodemographic data investigations and relevant clinical examinations. Lp(a) was analyzed by colorimetric enzymatic assays and hormone was measured with chemiluminescence immunoassay method. According to Lp(a) levels, they were categorized into 3 groups: the lower Lp(a) group (Lp(a) levels < 30 mg/dl), the moderate Lp(a) group (Lp(a) levels between 30 mg/dl and 120 mg/dl) and the higher Lp(a) group (Lp(a) levels > 120 mg/dl). The differences of hormone levels among the three groups were compared and the relationship between Lp(a) and hormones was analyzed by Spearman's rank correlation. RESULTS The higher Lp(a) group had significantly lower testosterone (TES) levels compared with the lower and moderate Lp(a) groups in the case of gender, age and BMI matching. Lp(a) concentration was negatively correlated with TES levels in all participants and the negative association between Lp(a) and TES levels was also observed when the analysis was stratified by gender. Additionally, the TES was statistically related with Lp(a) levels in the multiple linear regression model (95% confidence interval: - 0.451 to - 0.079). CONCLUSIONS TES levels was negatively associated with Lp(a) levels in Chinese overweight/obese patients.
Collapse
Affiliation(s)
- Xiaona Chang
- Department of Endocrinology, Beijing Chao-Yang Hospital, Capital Medical University, No. 8, Gongti South Road, Chaoyang District, Beijing, 100020, China
| | - Nannan Bian
- Department of Endocrinology, Beijing Chao-Yang Hospital, Capital Medical University, No. 8, Gongti South Road, Chaoyang District, Beijing, 100020, China
| | - Xiaoyu Ding
- Department of Endocrinology, Beijing Chao-Yang Hospital, Capital Medical University, No. 8, Gongti South Road, Chaoyang District, Beijing, 100020, China
| | - Jinman Li
- Department of Endocrinology, Beijing Chao-Yang Hospital, Capital Medical University, No. 8, Gongti South Road, Chaoyang District, Beijing, 100020, China
| | - Yu An
- Department of Endocrinology, Beijing Chao-Yang Hospital, Capital Medical University, No. 8, Gongti South Road, Chaoyang District, Beijing, 100020, China
| | - Jiaxuan Wang
- Department of Endocrinology, Beijing Chao-Yang Hospital, Capital Medical University, No. 8, Gongti South Road, Chaoyang District, Beijing, 100020, China
| | - Jia Liu
- Department of Endocrinology, Beijing Chao-Yang Hospital, Capital Medical University, No. 8, Gongti South Road, Chaoyang District, Beijing, 100020, China.
| | - Guang Wang
- Department of Endocrinology, Beijing Chao-Yang Hospital, Capital Medical University, No. 8, Gongti South Road, Chaoyang District, Beijing, 100020, China
| |
Collapse
|
28
|
Schwartz GG, Ballantyne CM. Existing and emerging strategies to lower Lipoprotein(a). Atherosclerosis 2022; 349:110-122. [DOI: 10.1016/j.atherosclerosis.2022.04.020] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/24/2021] [Revised: 04/13/2022] [Accepted: 04/16/2022] [Indexed: 12/24/2022]
|
29
|
Lamina C, Ward NC. Lipoprotein (a) and diabetes mellitus. Atherosclerosis 2022; 349:63-71. [DOI: 10.1016/j.atherosclerosis.2022.04.016] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/12/2021] [Revised: 04/13/2022] [Accepted: 04/14/2022] [Indexed: 12/24/2022]
|
30
|
Bustanji Y, Barham N, Abu-Rish EY, Alhyari A, Albustanji B, Alnajjar M, Abu-Irmaileh B, El-Huneidi W, Abu-Gharbieh E, Mohammad M, Fararjeh M, Issa A, Semreen M, Bajes H. Clinical investigation of lipoprotein (a) levels in type 2 diabetics for cariovascular diseases prediction and prognosis. Horm Mol Biol Clin Investig 2022; 43:263-271. [PMID: 35427448 DOI: 10.1515/hmbci-2021-0090] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Accepted: 03/13/2022] [Indexed: 11/15/2022]
Abstract
OBJECTIVES We aimed to evaluate the levels of serum lipoprotein a, LP (a), in Jordanian patients with type 2 diabetes mellitus (DM); and to examine its relation to glycemic control, metabolic syndrome (MS) and duration of DM. The LP (a) is considered one of the independent risk factors for coronary artery disease (CAD) in the general population. METHODS Fasting blood samples were drawn from 51 diabetic patients with type 2 DM and 31 non-diabetic age and sex control subjects. Serum LP (a) was measured along with other parameters, including triglycerides (TG), total cholesterol (TC), high-density lipoprotein cholesterol (HDL-c), low-density lipoprotein cholesterol (LDL-c) and glycosylated haemoglobin (HbA1c). Correlation analyses were performed between LP (a) and the various variables measured. RESULTS LP (a) measurement showed a skewed distribution towards the lower levels in both groups. Mean LP (a) levels showed a statistically insignificant difference between the two groups. No correlations of LP (a) were observed with age, sex or body mass index (BMI). No correlations of LP (a) with LDL-c, HDL-c, TG, TC, MS, DM duration or HbA1c were observed. The LP (a) serum levels were significantly higher in type 2 diabetic patients with retinopathy. CONCLUSIONS LP (a) serum levels are not increased in type 2 diabetic patients; so, LP (a) may not be a reliable marker for early therapeutic interventions in DM patients, even in high-risk for thrombosis groups.
Collapse
Affiliation(s)
- Yasser Bustanji
- Department of Basic Medical Sciences, College of Medicine, University of Sharjah, Sharjah, UAE.,School of Pharmacy, The University of Jordan, Amman, Jordan
| | - Noor Barham
- School of Pharmacy, The University of Jordan, Amman, Jordan
| | | | | | | | - Majd Alnajjar
- Hamdi Mango Center for Scientific Research, The University of Jordan, Amman, Jordan
| | - Bashaer Abu-Irmaileh
- Hamdi Mango Center for Scientific Research, The University of Jordan, Amman, Jordan
| | - Waseem El-Huneidi
- Department of Basic Medical Sciences, College of Medicine, University of Sharjah, Sharjah, UAE
| | - Eman Abu-Gharbieh
- Department of Basic Medical Sciences, College of Medicine, University of Sharjah, Sharjah, UAE
| | | | | | - Ala Issa
- School of Pharmacy, The University of Jordan, Amman, Jordan
| | | | - Hana Bajes
- School of Pharmacy, The University of Jordan, Amman, Jordan.,Science Department, Atlantic Cape Community College, Mays Landing, NJ 08330, USA
| |
Collapse
|
31
|
Diabetes and Familial Hypercholesterolemia: Interplay between Lipid and Glucose Metabolism. Nutrients 2022; 14:nu14071503. [PMID: 35406116 PMCID: PMC9002616 DOI: 10.3390/nu14071503] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Revised: 03/29/2022] [Accepted: 03/30/2022] [Indexed: 02/04/2023] Open
Abstract
Familial hypercholesterolemia (FH) is a genetic disease characterized by high low-density lipoprotein (LDL) cholesterol (LDL-c) concentrations that increase cardiovascular risk and cause premature death. The most frequent cause of the disease is a mutation in the LDL receptor (LDLR) gene. Diabetes is also associated with an increased risk of cardiovascular disease and mortality. People with FH seem to be protected from developing diabetes, whereas cholesterol-lowering treatments such as statins are associated with an increased risk of the disease. One of the hypotheses to explain this is based on the toxicity of LDL particles on insulin-secreting pancreatic β-cells, and their uptake by the latter, mediated by the LDLR. A healthy lifestyle and a relatively low body mass index in people with FH have also been proposed as explanations. Its association with superimposed diabetes modifies the phenotype of FH, both regarding the lipid profile and cardiovascular risk. However, findings regarding the association and interplay between these two diseases are conflicting. The present review summarizes the existing evidence and discusses knowledge gaps on the matter.
Collapse
|
32
|
Xia J, Guo C, Cao H, Liu K, Peng W, Sun Y, Xie Y, Li B, Zhang F, Wen F, Zhang L. Impact of lipoprotein(a) level on cardiometabolic disease in the Chinese population: The CHCN-BTH Study. Eur J Clin Invest 2022; 52:e13689. [PMID: 34632581 DOI: 10.1111/eci.13689] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Revised: 09/24/2021] [Accepted: 09/26/2021] [Indexed: 10/20/2022]
Abstract
BACKGROUND The emergence of promising compounds to lower lipoprotein(a) [Lp(a)] has increased the need for a precise characterisation and comparability assessment of Lp(a)-associated cardiometabolic disease risk. This study aimed to evaluate the distribution of Lp(a) levels in a Chinese population and characterise the association with cardiometabolic diseases. METHODS We assessed data from individuals from the Cohort Study on Chronic Diseases of the General Community Population in the Beijing-Tianjin-Hebei Region project. All Lp(a) measurements were performed in the same hospital. The cardiometabolic diseases considered were coronary heart disease (CHD), stroke, hypertension and type 2 diabetes (T2DM). RESULTS A total of 25343 individuals were included in the study. The median level of Lp(a) was 11.9 mg/dl (IQR 5.9 to 23.7 mg/dl), and higher Lp(a) levels showed a significant concentration-dependent association with CHD risk. Individuals with Lp(a) levels lower than the 25th percentile were at increased risk of hypertension (OR: 1.15, 95% CI: 1.06-1.25) and T2DM (OR: 1.15, 95% CI: 1.03-1.28); however, Lp(a) levels were not significantly associated with stroke. The addition of Lp(a) levels to the prognostic model led to a marginal but significant C-index, integrated discrimination improvement and net reclassification improvement. CONCLUSIONS In this large sample size study, we observed that elevated Lp(a) levels were significantly associated with CHD. Furthermore, we found that the lowest Lp(a) levels were also significantly associated with hypertension and T2DM. These results provide evidence for differential approaches to higher levels of Lp(a) in individuals with different cardiometabolic diseases.
Collapse
Affiliation(s)
- Juan Xia
- Department of Epidemiology and Health Statistics, School of Public Health, Capital Medical University, and Beijing Municipal Key Laboratory of Clinical Epidemiology, Beijing, China
| | - Chunyue Guo
- Department of Epidemiology and Health Statistics, School of Public Health, Capital Medical University, and Beijing Municipal Key Laboratory of Clinical Epidemiology, Beijing, China
| | - Han Cao
- Department of Epidemiology and Health Statistics, School of Public Health, Capital Medical University, and Beijing Municipal Key Laboratory of Clinical Epidemiology, Beijing, China
| | - Kuo Liu
- Department of Epidemiology and Health Statistics, School of Public Health, Capital Medical University, and Beijing Municipal Key Laboratory of Clinical Epidemiology, Beijing, China
| | - Wenjuan Peng
- Department of Epidemiology and Health Statistics, School of Public Health, Capital Medical University, and Beijing Municipal Key Laboratory of Clinical Epidemiology, Beijing, China
| | - Yanyan Sun
- Department of Epidemiology and Health Statistics, School of Public Health, Capital Medical University, and Beijing Municipal Key Laboratory of Clinical Epidemiology, Beijing, China
| | - Yunyi Xie
- Department of Epidemiology and Health Statistics, School of Public Health, Capital Medical University, and Beijing Municipal Key Laboratory of Clinical Epidemiology, Beijing, China
| | - Bingxiao Li
- Department of Epidemiology and Health Statistics, School of Public Health, Capital Medical University, and Beijing Municipal Key Laboratory of Clinical Epidemiology, Beijing, China
| | - Fengxu Zhang
- Department of Epidemiology and Health Statistics, School of Public Health, Capital Medical University, and Beijing Municipal Key Laboratory of Clinical Epidemiology, Beijing, China
| | - Fuyuan Wen
- Department of Epidemiology and Health Statistics, School of Public Health, Capital Medical University, and Beijing Municipal Key Laboratory of Clinical Epidemiology, Beijing, China
| | - Ling Zhang
- Department of Epidemiology and Health Statistics, School of Public Health, Capital Medical University, and Beijing Municipal Key Laboratory of Clinical Epidemiology, Beijing, China
| |
Collapse
|
33
|
Wang J, Sun H, Wang Y, An Y, Liu J, Wang G. Glucose metabolism status modifies the relationship between lipoprotein(a) and carotid plaques in individuals with fatty liver disease. Front Endocrinol (Lausanne) 2022; 13:947914. [PMID: 36465632 PMCID: PMC9709428 DOI: 10.3389/fendo.2022.947914] [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: 05/19/2022] [Accepted: 10/31/2022] [Indexed: 11/17/2022] Open
Abstract
BACKGROUND AND AIMS Glucose and lipoprotein(a) [Lp(a)] have been recognized risk factors for atherosclerosis. The impact of both factors on fatty liver patients has not been studied. The aim of this study is to explore the role of high-level Lp(a) and different glucose metabolism statuses on carotid plaques in fatty liver patients. METHODS We selected 4,335 fatty liver patients in this cross-sectional study. The diagnosis of fatty liver disease and carotid plaques was made by ultrasound. Participants were divided into four groups based on glucose metabolism status (normal glucose regulation [NGR], lower bound of impaired fasting glucose [IFG-L], higher bound of impaired fasting glucose [IFG-H], diabetes mellitus [DM]) and then categorized into 12 subgroups according to Lp(a) concentrations. The association between variables was estimated by odds ratio (OR). RESULTS Carotid plaques were present in 1,613 (37.2%) fatty liver patients. Lp(a)≥30 mg/dL was associated with high risk of carotid plaques in those patients with IFG-L, IFG-H and DM (OR 1.934 [95% CI 1.033-3.618], 2.667 [1.378-5.162], 4.000 [2.219-7.210], respectively; p<0.05). Fatty liver patients with DM plus Lp(a)<10 mg/dL and 10≤Lp(a)<30 mg/dL were more vulnerable to carotid plaques (OR 1.563 [95% CI 1.090-2.241], 1.930 [1.279-2.914]), respectively, p<0.05). CONCLUSIONS Our study first suggested that high-level Lp(a) may raise the risk of carotid plaques in fatty liver patients with not only diabetes but also IFG, manifesting that Lp(a) may be helpful for the early discovery of subclinical atherosclerosis in fatty liver patients with impaired glucose metabolism.
Collapse
Affiliation(s)
- Jiaxuan Wang
- Department of Endocrinology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Honglin Sun
- Department of Endocrinology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Ying Wang
- Physical Examination Center, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Yu An
- Department of Endocrinology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Jia Liu
- Department of Endocrinology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Guang Wang
- Department of Endocrinology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
- *Correspondence: Guang Wang,
| |
Collapse
|
34
|
Durlach V, Bonnefont-Rousselot D, Boccara F, Varret M, Di-Filippo Charcosset M, Cariou B, Valero R, Charriere S, Farnier M, Morange PE, Meilhac O, Lambert G, Moulin P, Gillery P, Beliard-Lasserre S, Bruckert E, Carrié A, Ferrières J, Collet X, Chapman MJ, Anglés-Cano E. Lipoprotein(a): Pathophysiology, measurement, indication and treatment in cardiovascular disease. A consensus statement from the Nouvelle Société Francophone d'Athérosclérose (NSFA). Arch Cardiovasc Dis 2021; 114:828-847. [PMID: 34840125 DOI: 10.1016/j.acvd.2021.10.009] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Revised: 10/16/2021] [Accepted: 10/18/2021] [Indexed: 10/19/2022]
Abstract
Lipoprotein(a) is an apolipoprotein B100-containing low-density lipoprotein-like particle that is rich in cholesterol, and is associated with a second major protein, apolipoprotein(a). Apolipoprotein(a) possesses structural similarity to plasminogen but lacks fibrinolytic activity. As a consequence of its composite structure, lipoprotein(a) may: (1) elicit a prothrombotic/antifibrinolytic action favouring clot stability; and (2) enhance atherosclerosis progression via its propensity for retention in the arterial intima, with deposition of its cholesterol load at sites of plaque formation. Equally, lipoprotein(a) may induce inflammation and calcification in the aortic leaflet valve interstitium, leading to calcific aortic valve stenosis. Experimental, epidemiological and genetic evidence support the contention that elevated concentrations of lipoprotein(a) are causally related to atherothrombotic risk and equally to calcific aortic valve stenosis. The plasma concentration of lipoprotein(a) is principally determined by genetic factors, is not influenced by dietary habits, remains essentially constant over the lifetime of a given individual and is the most powerful variable for prediction of lipoprotein(a)-associated cardiovascular risk. However, major interindividual variations (up to 1000-fold) are characteristic of lipoprotein(a) concentrations. In this context, lipoprotein(a) assays, although currently insufficiently standardized, are of considerable interest, not only in stratifying cardiovascular risk, but equally in the clinical follow-up of patients treated with novel lipid-lowering therapies targeted at lipoprotein(a) (e.g. antiapolipoprotein(a) antisense oligonucleotides and small interfering ribonucleic acids) that markedly reduce circulating lipoprotein(a) concentrations. We recommend that lipoprotein(a) be measured once in subjects at high cardiovascular risk with premature coronary heart disease, in familial hypercholesterolaemia, in those with a family history of coronary heart disease and in those with recurrent coronary heart disease despite lipid-lowering treatment. Because of its clinical relevance, the cost of lipoprotein(a) testing should be covered by social security and health authorities.
Collapse
Affiliation(s)
- Vincent Durlach
- Champagne-Ardenne University, UMR CNRS 7369 MEDyC & Cardio-Thoracic Department, Reims University Hospital, 51092 Reims, France
| | - Dominique Bonnefont-Rousselot
- Metabolic Biochemistry Department, Hôpital Pitié-Salpêtrière, AP-HP, 75013 Paris, France; Université de Paris, CNRS, INSERM, UTCBS, 75006 Paris, France
| | - Franck Boccara
- Sorbonne University, GRC n(o) 22, C(2)MV, INSERM UMR_S 938, Centre de Recherche Saint-Antoine, IHU ICAN, 75012 Paris, France; Service de Cardiologie, Hôpital Saint-Antoine, AP-HP, 75012 Paris, France
| | - Mathilde Varret
- Laboratory for Vascular Translational Science (LVTS), INSERM U1148, Centre Hospitalier Universitaire Xavier Bichat, 75018 Paris, France; Université de Paris, 75018 Paris, France
| | - Mathilde Di-Filippo Charcosset
- Hospices Civils de Lyon, UF Dyslipidémies, 69677 Bron, France; Laboratoire CarMen, INSERM, INRA, INSA, Université Claude-Bernard Lyon 1, 69495 Pierre-Bénite, France
| | - Bertrand Cariou
- Université de Nantes, CHU Nantes, CNRS, INSERM, l'Institut du Thorax, 44000 Nantes, France
| | - René Valero
- Endocrinology Department, La Conception Hospital, AP-HM, Aix-Marseille University, INSERM, INRAE, C2VN, 13005 Marseille, France
| | - Sybil Charriere
- Hospices Civils de Lyon, INSERM U1060, Laboratoire CarMeN, Université Lyon 1, 69310 Pierre-Bénite, France
| | - Michel Farnier
- PEC2, EA 7460, University of Bourgogne Franche-Comté, 21079 Dijon, France; Department of Cardiology, CHU Dijon Bourgogne, 21000 Dijon, France
| | - Pierre E Morange
- Aix-Marseille University, INSERM, INRAE, C2VN, 13385 Marseille, France
| | - Olivier Meilhac
- INSERM, UMR 1188 DéTROI, Université de La Réunion, 97744 Saint-Denis de La Réunion, Reunion; CHU de La Réunion, CIC-EC 1410, 97448 Saint-Pierre, Reunion
| | - Gilles Lambert
- INSERM, UMR 1188 DéTROI, Université de La Réunion, 97744 Saint-Denis de La Réunion, Reunion; CHU de La Réunion, CIC-EC 1410, 97448 Saint-Pierre, Reunion
| | - Philippe Moulin
- Hospices Civils de Lyon, INSERM U1060, Laboratoire CarMeN, Université Lyon 1, 69310 Pierre-Bénite, France
| | - Philippe Gillery
- Laboratory of Biochemistry-Pharmacology-Toxicology, Reims University Hospital, University of Reims Champagne-Ardenne, UMR CNRS/URCA n(o) 7369, 51092 Reims, France
| | - Sophie Beliard-Lasserre
- Endocrinology Department, La Conception Hospital, AP-HM, Aix-Marseille University, INSERM, INRAE, C2VN, 13005 Marseille, France
| | - Eric Bruckert
- Service d'Endocrinologie-Métabolisme, Hôpital Pitié-Salpêtrière, AP-HP, 75013 Paris, France; IHU ICAN, Sorbonne University, 75013 Paris, France
| | - Alain Carrié
- Sorbonne University, UMR INSERM 1166, IHU ICAN, Laboratory of Endocrine and Oncological Biochemistry, Obesity and Dyslipidaemia Genetic Unit, Hôpital Pitié-Salpêtrière, AP-HP, 75013 Paris, France
| | - Jean Ferrières
- Department of Cardiology and INSERM UMR 1295, Rangueil University Hospital, TSA 50032, 31059 Toulouse, France
| | - Xavier Collet
- INSERM U1048, Institute of Metabolic and Cardiovascular Diseases, Rangueil University Hospital, BP 84225, 31432 Toulouse, France
| | - M John Chapman
- Sorbonne University, Hôpital Pitié-Salpêtrière and National Institute for Health and Medical Research (INSERM), 75013 Paris, France
| | - Eduardo Anglés-Cano
- Université de Paris, INSERM, Innovative Therapies in Haemostasis, 75006 Paris, France.
| |
Collapse
|
35
|
Pluimakers VG, van Santen SS, Fiocco M, Bakker MCE, van der Lelij AJ, van den Heuvel-Eibrink MM, Neggers SJCMM. Can biomarkers be used to improve diagnosis and prediction of metabolic syndrome in childhood cancer survivors? A systematic review. Obes Rev 2021; 22:e13312. [PMID: 34258851 PMCID: PMC8596408 DOI: 10.1111/obr.13312] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Revised: 05/11/2021] [Accepted: 06/09/2021] [Indexed: 12/26/2022]
Abstract
Childhood cancer survivors (CCS) are at increased risk to develop metabolic syndrome (MetS), diabetes, and cardiovascular disease. Common criteria underestimate adiposity and possibly underdiagnose MetS, particularly after abdominal radiotherapy. A systematic literature review and meta-analysis on the diagnostic and predictive value of nine newer MetS related biomarkers (adiponectin, leptin, uric acid, hsCRP, TNF-alpha, IL-1, IL-6, apolipoprotein B (apoB), and lipoprotein(a) [lp(a)]) in survivors and adult non-cancer survivors was performed by searching PubMed and Embase. Evidence was summarized with GRADE after risk of bias evaluation (QUADAS-2/QUIPS). Eligible studies on promising biomarkers were pooled. We identified 175 general population and five CCS studies. In the general population, valuable predictive biomarkers are uric acid, adiponectin, hsCRP and apoB (high level of evidence), and leptin (moderate level of evidence). Valuable diagnostic biomarkers are hsCRP, adiponectin, uric acid, and leptin (low, low, moderate, and high level of evidence, respectively). Meta-analysis showed OR for hyperuricemia of 2.94 (age-/sex-adjusted), OR per unit uric acid increase of 1.086 (unadjusted), and AUC for hsCRP of 0.71 (unadjusted). Uric acid, adiponectin, hsCRP, leptin, and apoB can be alternative biomarkers in the screening setting for MetS in survivors, to enhance early identification of those at high risk of subsequent complications.
Collapse
Affiliation(s)
| | - Selveta S van Santen
- Princess Máxima Center for Pediatric Oncology, Utrecht, Netherlands.,Department of Medicine, Endocrinology, Erasmus Medical Center, Rotterdam, Netherlands
| | - Marta Fiocco
- Princess Máxima Center for Pediatric Oncology, Utrecht, Netherlands.,Medical Statistics, Department of Biomedical Data Science, Leiden UMC, Leiden, Netherlands.,Mathematical Institute, Leiden University, Leiden, Netherlands
| | - Marie-Christine E Bakker
- Princess Máxima Center for Pediatric Oncology, Utrecht, Netherlands.,Department of Medicine, University Medical Center Utrecht, Netherlands
| | - Aart J van der Lelij
- Department of Medicine, Endocrinology, Erasmus Medical Center, Rotterdam, Netherlands
| | | | - Sebastian J C M M Neggers
- Princess Máxima Center for Pediatric Oncology, Utrecht, Netherlands.,Department of Medicine, Endocrinology, Erasmus Medical Center, Rotterdam, Netherlands
| |
Collapse
|
36
|
Lipoprotein (a) and Cardiovascular Disease: A Missing Link for Premature Atherosclerotic Heart Disease and/or Residual Risk. J Cardiovasc Pharmacol 2021; 79:e18-e35. [PMID: 34694242 DOI: 10.1097/fjc.0000000000001160] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Accepted: 09/30/2021] [Indexed: 11/26/2022]
Abstract
ABSTRACT Lipoprotein(a) or lipoprotein "little a" is an under-recognized causal risk factor for cardiovascular (CV) disease (CVD), including coronary atherosclerosis, aortic valvular stenosis, ischemic stroke, heart failure and peripheral arterial disease. Elevated plasma Lp(a) (≥50 mg/dL or ≥100 nmol/L) is commonly encountered in almost 1 in 5 individuals and confers a higher CV risk compared to those with normal Lp(a) levels, although such normal levels have not been generally agreed upon. Elevated Lp(a) is considered a cause of premature and accelerated atherosclerotic CVD. Thus, in patients with a positive family or personal history of premature coronary artery disease (CAD), Lp(a) should be measured. However, elevated Lp(a) may confer increased risk for incident CAD even in the absence of a family history of CAD, and even in those who have guideline-lowered LDL-cholesterol (<70 mg/dl) and continue to have a persisting CV residual risk. Thus, measurement of Lp(a) will have a significant clinical impact on the assessment of atherosclerotic CVD risk, and will assume a more important role in managing patients with CVD with the advent and clinical application of specific Lp(a)-lowering therapies. Conventional therapeutic approaches like lifestyle modification and statin therapy remain ineffective at lowering Lp(a). Newer treatment modalities, such as gene silencing via RNA interference with use of antisense oligonucleotide(s) or small interfering RNA molecules targeting Lp(a) seem very promising. These issues are herein reviewed, accumulated data are scrutinized, meta-analyses and current guidelines are tabulated and Lp(a)-related CVDs and newer therapeutic modalities are pictorially illustrated.
Collapse
|
37
|
Ruscica M, Sirtori CR, Corsini A, Watts GF, Sahebkar A. Lipoprotein(a): Knowns, unknowns and uncertainties. Pharmacol Res 2021; 173:105812. [PMID: 34450317 DOI: 10.1016/j.phrs.2021.105812] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Revised: 08/11/2021] [Accepted: 08/11/2021] [Indexed: 02/07/2023]
Abstract
Over the last 10 years, there have been advances on several aspects of lipoprotein(a) which are reviewed in the present article. Since the standard immunoassays for measuring lipoprotein(a) are not fully apo(a) isoform-insensitive, the application of an LC-MS/MS method for assaying molar concentrations of lipoprotein(a) has been advocated. Genome wide association, epidemiological, and clinical studies have established high lipoprotein(a) as a causal risk factor for atherosclerotic cardiovascular diseases (ASCVD). However, the relative importance of molar concentration, apo(a) isoform size or variants within the LPA gene is still controversial. Lipoprotein(a)-raising single nucleotide polymorphisms has not been shown to add on value in predicting ASCVD beyond lipoprotein(a) concentrations. Although hyperlipoproteinemia(a) represents an important confounder in the diagnosis of familial hypercholesterolemia (FH), it enhances the risk of ASCVD in these patients. Thus, identification of new cases of hyperlipoproteinemia(a) during cascade testing can increase the identification of high-risk individuals. However, it remains unclear whether FH itself increases lipoprotein(a). The ASCVD risk associated with lipoprotein(a) seems to follow a linear gradient across the distribution, regardless of racial subgroups and other risk factors. The inverse association with the risk of developing type 2 diabetes needs consideration as effective lipoprotein(a) lowering therapies are progressing towards the market. Considering that Mendelian randomization analyses have identified the degree of lipoprotein(a)-lowering that is required to achieve ASCVD benefit, the findings of the ongoing outcome trial with pelacarsen will clarify whether dramatically lowering lipoprotein(a) levels can reduce the risk of ASCVD.
Collapse
Affiliation(s)
- Massimiliano Ruscica
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Italy.
| | - Cesare R Sirtori
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Italy
| | - Alberto Corsini
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Italy; IRCCS MultiMedica, Sesto S. Giovanni, Milan, Italy
| | - Gerald F Watts
- School of Medicine, University of Western Australia, Perth, Australia; Lipid Disorders Clinic, Cardiometabolic Services, Department of Cardiology, Royal Perth Hospital, Australia
| | - Amirhossein Sahebkar
- School of Medicine, University of Western Australia, Perth, Australia; Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran; Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
38
|
Satterfield BA, Dikilitas O, Safarova MS, Clarke SL, Tcheandjieu C, Zhu X, Bastarache L, Larson EB, Justice AE, Shang N, Rosenthal EA, Shah AS, Namjou-Khales B, Urbina EM, Wei WQ, Feng Q, Jarvik GP, Hebbring SJ, de Andrade M, Manolio TA, Assimes TL, Kullo IJ. Associations of Genetically Predicted Lp(a) (Lipoprotein [a]) Levels With Cardiovascular Traits in Individuals of European and African Ancestry. CIRCULATION. GENOMIC AND PRECISION MEDICINE 2021; 14:e003354. [PMID: 34282949 PMCID: PMC8634549 DOI: 10.1161/circgen.120.003354] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
BACKGROUND Lp(a) (lipoprotein [a]) levels are higher in individuals of African ancestry (AA) than in individuals of European ancestry (EA). We examined associations of genetically predicted Lp(a) levels with (1) atherosclerotic cardiovascular disease subtypes: coronary heart disease, cerebrovascular disease, peripheral artery disease, and abdominal aortic aneurysm and (2) nonatherosclerotic cardiovascular disease phenotypes, stratified by ancestry. METHODS We performed (1) Mendelian randomization analyses for previously reported cardiovascular associations and (2) Mendelian randomization-phenome-wide association analyses for novel associations. Analyses were stratified by ancestry in electronic Medical Records and Genomics, United Kingdom Biobank, and Million Veteran Program cohorts separately and in a combined cohort of 804 507 EA and 103 580 AA participants. RESULTS In Mendelian randomization analyses using the combined cohort, a 1-SD genetic increase in Lp(a) level was associated with atherosclerotic cardiovascular disease subtypes in EA-odds ratio and 95% CI for coronary heart disease 1.28 (1.16-1.41); cerebrovascular disease 1.14 (1.07-1.21); peripheral artery disease 1.22 (1.11-1.34); abdominal aortic aneurysm 1.28 (1.17-1.40); in AA, the effect estimate was lower than in EA and nonsignificant for coronary heart disease 1.11 (0.99-1.24) and cerebrovascular disease 1.06 (0.99-1.14) but similar for peripheral artery disease 1.16 (1.01-1.33) and abdominal aortic aneurysm 1.34 (1.11-1.62). In EA, a 1-SD genetic increase in Lp(a) level was associated with aortic valve disorders 1.34 (1.10-1.62), mitral valve disorders 1.18 (1.09-1.27), congestive heart failure 1.12 (1.05-1.19), and chronic kidney disease 1.07 (1.01-1.14). In AA, no significant associations were noted for aortic valve disorders 1.08 (0.94-1.25), mitral valve disorders 1.02 (0.89-1.16), congestive heart failure 1.02 (0.95-1.10), or chronic kidney disease 1.05 (0.99-1.12). Mendelian randomization-phenome-wide association analyses identified novel associations in EA with arterial thromboembolic disease, nonaortic aneurysmal disease, atrial fibrillation, cardiac conduction disorders, and hypertension. CONCLUSIONS Many cardiovascular associations of genetically increased Lp(a) that were significant in EA were not significant in AA. Lp(a) was associated with atherosclerotic cardiovascular disease in four major arterial beds in EA but only with peripheral artery disease and abdominal aortic aneurysm in AA. Additionally, novel cardiovascular associations were detected in EA.
Collapse
Affiliation(s)
| | - Ozan Dikilitas
- Dept of Cardiovascular Medicine, Mayo Clinic, Rochester, MN
| | | | - Shoa L. Clarke
- VA Palo Alto Health Care System, Palo Alto
- Division of Cardiovascular Medicine, Dept of Medicine, Stanford Univ School of Medicine, Stanford, CA
| | - Catherine Tcheandjieu
- VA Palo Alto Health Care System, Palo Alto
- Division of Cardiovascular Medicine, Dept of Medicine, Stanford Univ School of Medicine, Stanford, CA
- Dept of Pediatric Cardiology, Stanford Univ School of Medicine, Stanford, CA
| | - Xiang Zhu
- VA Palo Alto Health Care System, Palo Alto
- Dept of Statistics, The Pennsylvania State Univ, University Park, PA
- Huck Institutes of the Life Sciences, The Pennsylvania State Univ, University Park, PA
- Dept of Statistics, Stanford Univ, Stanford, CA
| | - Lisa Bastarache
- Dept of Biomedical Informatics, Vanderbilt Univ, Nashville, TN
| | - Eric B. Larson
- Kaiser Permanente Washington Health Research Institutes, Seattle, WA
| | | | - Ning Shang
- Dept of Biomedical Informatics, Columbia Univ, New York, NY
| | | | - Amy Sanghavi Shah
- Division of Endocrinology, Cincinnati Children’s Hospital Medical Center & Univ of Cincinnati
| | - Bahram Namjou-Khales
- Center for Autoimmune Genomics and Etiology, Cincinnati Children’s Hospital Medical Center & Dept of Pediatrics, Univ of Cincinnati, College of Medicine, Cincinnati, OH
| | - Elaine M. Urbina
- Division of Endocrinology, Heart Institute, Cincinnati Children’s Hospital Medical Center & Univ of Cincinnati
| | - Wei-Qi Wei
- Dept of Biomedical Informatics, Vanderbilt Univ, Nashville, TN
| | - QiPing Feng
- Division of Clinical Pharmacology, Dept of Medicine, Vanderbilt Univ Medical Center, Nashville, TN
| | - Gail P. Jarvik
- Division of Medical Genetics, Dept of Medicine, Univ of Washington, Seattle, WA
| | - Scott J. Hebbring
- Center for Precision Medicine, Marshfield Clinic Research Institute, WI
| | - Mariza de Andrade
- Dept of Cardiovascular Medicine, Dept of Health Sciences Research, Mayo Clinic, Rochester, MN
| | - Teri A. Manolio
- Division of Genomic Medicine, National Human Genome Research Institute, Bethesda, MD
| | | | - Iftikhar J. Kullo
- Dept of Cardiovascular Medicine, Mayo Clinic, Rochester, MN
- Dept of Cardiovascular Medicine, Gonda Vascular Center, Mayo Clinic, Rochester, MN
| |
Collapse
|
39
|
Langsted A, Nordestgaard BG. Genetics of Lipoprotein(a): Cardiovascular Disease and Future Therapy. Curr Atheroscler Rep 2021; 23:46. [PMID: 34148150 DOI: 10.1007/s11883-021-00937-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/17/2021] [Indexed: 12/14/2022]
Abstract
PURPOSE OF REVIEW Lipoprotein(a) levels are determined 80-90% by genetics and differ by up to 1000-fold between individuals. This review discusses the most recent literature on lipoprotein(a) as a risk factor for cardiovascular disease, as well as future lipoprotein(a)lowering therapies. RECENT FINDINGS Over the past few decades, numerous studies have observed that high lipoprotein(a) levels are associated observationally and causally through human genetics with increased risk of cardiovascular disease. Also, the development of safe and effective therapies to lower lipoprotein(a) is ongoing, most importantly using antisense oligonucleotides to prevent production of lipoprotein(a). Finally, both observational and genetic studies have estimated the extent to which lowering of lipoprotein(a) is needed to obtain a clinically meaningful reduction in the risk of cardiovascular disease. Lipoprotein(a) is a causal risk factor for cardiovascular disease; however, currently no approved safe and effective therapy is available to lower lipoprotein(a) levels. That said, promising randomized studies using antisense oligonucleotides show up to 80% reductions in lipoprotein(a), reductions that hopefully will result in lowering the risk of cardiovascular disease as presently tested in the ongoing HORIZON phase 3 trial.
Collapse
Affiliation(s)
- Anne Langsted
- Department of Clinical Biochemistry, Herlev and Gentofte Hospital, Copenhagen University Hospital, Borgmester Ib Juuls Vej 73, Entrance 7, 4th floor, N5, DK-2730, Herlev, Denmark. .,The Copenhagen General Population Study, Herlev and Gentofte Hospital, Copenhagen University Hospital, Borgmester Ib Juuls Vej 73, Entrance 7, 4th floor, N5, DK-2730, Herlev, Denmark. .,Department of Clinical Medicine Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.
| | - Børge G Nordestgaard
- Department of Clinical Biochemistry, Herlev and Gentofte Hospital, Copenhagen University Hospital, Borgmester Ib Juuls Vej 73, Entrance 7, 4th floor, N5, DK-2730, Herlev, Denmark.,The Copenhagen General Population Study, Herlev and Gentofte Hospital, Copenhagen University Hospital, Borgmester Ib Juuls Vej 73, Entrance 7, 4th floor, N5, DK-2730, Herlev, Denmark.,Department of Clinical Medicine Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
40
|
Schwartz GG, Szarek M, Bittner VA, Bhatt DL, Diaz R, Goodman SG, Jukema JW, Loy M, Manvelian G, Pordy R, White HD, Steg PG. Relation of Lipoprotein(a) Levels to Incident Type 2 Diabetes and Modification by Alirocumab Treatment. Diabetes Care 2021; 44:1219-1227. [PMID: 33722880 PMCID: PMC8132323 DOI: 10.2337/dc20-2842] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Accepted: 02/08/2021] [Indexed: 02/03/2023]
Abstract
OBJECTIVE In observational data, lower levels of lipoprotein(a) have been associated with greater prevalence of type 2 diabetes. Whether pharmacologic lowering of lipoprotein(a) influences incident type 2 diabetes is unknown. We determined the relationship of lipoprotein(a) concentration with incident type 2 diabetes and effects of treatment with alirocumab, a PCSK9 inhibitor. RESEARCH DESIGN AND METHODS In the ODYSSEY OUTCOMES trial alirocumab was compared with placebo in patients with acute coronary syndrome. Incident diabetes was determined from laboratory, medication, and adverse event data. RESULTS Among 13,480 patients without diabetes at baseline, 1,324 developed type 2 diabetes over a median 2.7 years. Median baseline lipoprotein(a) was 21.9 mg/dL. With placebo, 10 mg/dL lower baseline lipoprotein(a) was associated with hazard ratio 1.04 (95% CI 1.02-1.06, P < 0.001) for incident type 2 diabetes. Alirocumab reduced lipoprotein(a) by a median 23.2% with greater absolute reductions from higher baseline levels and no overall effect on incident type 2 diabetes (hazard ratio 0.95, 95% CI 0.85-1.05). At low baseline lipoprotein(a) levels, alirocumab tended to reduce incident type 2 diabetes, while at high baseline lipoprotein(a) alirocumab tended to increase incident type 2 diabetes compared with placebo (treatment-baseline lipoprotein(a) interaction P = 0.006). In the alirocumab group, a 10 mg/dL decrease in lipoprotein(a) from baseline was associated with hazard ratio 1.07 (95% CI 1.03-1.12; P = 0.0002) for incident type 2 diabetes. CONCLUSIONS In patients with acute coronary syndrome, baseline lipoprotein(a) concentration associated inversely with incident type 2 diabetes. Alirocumab had neutral overall effect on incident type 2 diabetes. However, treatment-related reductions in lipoprotein(a), more pronounced from high baseline levels, were associated with increased risk of incident type 2 diabetes. Whether these findings pertain to other therapies that reduce lipoprotein(a) is undetermined.
Collapse
Affiliation(s)
- Gregory G Schwartz
- Division of Cardiology, University of Colorado School of Medicine, Aurora, CO
| | - Michael Szarek
- Division of Cardiology, University of Colorado School of Medicine, Aurora, CO.,CPC Clinical Research, Aurora, CO.,Downstate School of Public Health, State University of New York, Brooklyn, NY
| | | | - Deepak L Bhatt
- Brigham and Women's Hospital and Harvard Medical School, Boston, MA
| | - Rafael Diaz
- Estudios Cardiológicos Latinoamérica, Instituto Cardiovascular de Rosario, Rosario, Argentina
| | - Shaun G Goodman
- Canadian VIGOUR Centre, University of Alberta, Edmonton, Alberta, Canada.,St. Michael's Hospital, University of Toronto, Toronto, Ontario, Canada
| | | | | | | | | | - Harvey D White
- Green Lane Cardiovascular Services, Auckland City Hospital, Auckland, New Zealand
| | | | | |
Collapse
|
41
|
Page MM, Watts GF. Contemporary perspectives on the genetics and clinical use of lipoprotein(a) in preventive cardiology. Curr Opin Cardiol 2021; 36:272-280. [PMID: 33741767 DOI: 10.1097/hco.0000000000000842] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
PURPOSE OF REVIEW The pathogenicity of lipoprotein(a) [Lp(a)] as a risk factor for atherosclerotic cardiovascular disease (ASCVD) is well evidenced and recognized by international consensus-based guidelines. However, the measurement of Lp(a) is not routine clinical practice. Therapeutic agents targeting Lp(a) are now progressing through randomised clinical trials, and it is timely for clinicians to familiarize themselves with this complex and enigmatic lipoprotein particle. RECENT FINDINGS Recent developments in the understanding of genetic influences on the structure, plasma concentration and atherogenicity of Lp(a) have contextualized its clinical relevance. Mendelian randomization studies have enabled estimation of the contribution of Lp(a) to ASCVD risk. Genotyping individual patients with respect to Lp(a)-raising single nucleotide polymorphisms predicts ASCVD, but has not yet been shown to add value beyond the measurement of Lp(a) plasma concentrations, which should be done by Lp(a) isoform-independent assays capable of reporting in molar concentrations. Contemporary gene-silencing technology underpins small interfering RNA and antisense oligonucleotides, which are emerging as the leading Lp(a)-lowering therapeutic agents. The degree of Lp(a)-lowering required to achieve meaningful reductions in ASCVD risk has been estimated by Mendelian randomization, providing conceptual support. SUMMARY Measurement of Lp(a) in the clinical setting contributes to the assessment of ASCVD risk, and will become more important with the advent of specific Lp(a)-lowering therapies. Knowledge of an individual patient's genetic predisposition to increased Lp(a) appears to impart little or not additional clinical value beyond Lp(a) particle concentration.
Collapse
Affiliation(s)
- Michael M Page
- School of Medicine, University of Western Australia, Crawley
- Western Diagnostic Pathology
| | - Gerald F Watts
- School of Medicine, University of Western Australia, Crawley
- Lipid Disorders Clinic, Cardiovascular Medicine, Royal Perth Hospital, Perth, Western Australia, Australia
| |
Collapse
|
42
|
Lan NSR, Burns K, Bell DA, Watts GF. Best practice for treating dyslipidaemia in patients with diabetes based on current international guidelines. Curr Opin Endocrinol Diabetes Obes 2021; 28:104-113. [PMID: 33278128 DOI: 10.1097/med.0000000000000594] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
PURPOSE OF REVIEW Dyslipidaemia is a major modifiable risk factor for atherosclerotic cardiovascular disease (ASCVD) in type 2 diabetes. We provide an in-context overview of recent trials of lipid-lowering pharmacotherapies and of recommendations from international guidelines for managing dyslipidaemia in patients with diabetes. RECENT FINDINGS Clinical trials have demonstrated that patients with diabetes derive greater benefits from ezetimibe and proprotein convertase subtilisin-kexin type 9 inhibitors owing to the higher absolute ASCVD risk compared with patients without diabetes. Pure eicosapentaenoic acid ethyl ester therapy should be considered in high risk patients with diabetes and hypertriglyceridaemia who have well controlled low-density lipoprotein cholesterol on statin therapy. International guidelines from USA, Canada and Europe have been updated to support a more intensive approach to treating dyslipidaemia in diabetes. SUMMARY Dyslipidaemia should be identified and treated intensively as part of overall diabetes management to reduce ASCVD risk. Although lifestyle modifications and statin therapy remain the cornerstone of management, add-on therapies should be strongly considered depending on the absolute risk of ASCVD and the degree of dyslipidaemia.
Collapse
Affiliation(s)
- Nick S R Lan
- Departments of Internal Medicine and Cardiology, Royal Perth Hospital
| | - Kharis Burns
- Medical School, University of Western Australia
- Department of Endocrinology and Diabetes, Royal Perth Hospital
| | - Damon A Bell
- Departments of Internal Medicine and Cardiology, Royal Perth Hospital
- Medical School, University of Western Australia
- Department of Clinical Biochemistry, PathWest Laboratory Medicine WA, Royal Perth and Fiona Stanley Hospitals
- Department of Biochemistry, Clinipath Pathology, Perth, Western Australia, Australia
| | - Gerald F Watts
- Departments of Internal Medicine and Cardiology, Royal Perth Hospital
- Medical School, University of Western Australia
| |
Collapse
|
43
|
Ward NC, Vickneswaran S, Watts GF. Lipoprotein (a) and diabetes mellitus: causes and consequences. Curr Opin Endocrinol Diabetes Obes 2021; 28:181-187. [PMID: 33229929 DOI: 10.1097/med.0000000000000597] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
PURPOSE OF REVIEW This review provides an update on the role of lipoprotein (a) [Lp(a)] in diabetes, including its impact as a risk factor as well as its contribution to the development of cardiovascular disease. RECENT FINDINGS Although a specific role for Lp(a) has not yet been conclusively established, it appears to have an inverse association with risk of diabetes. Several population-based studies have demonstrated associations between low levels of Lp(a) and increased risk of type 2 diabetes, but Mendelian randomization studies do not consistently support causality. Conversely, in patients with type 2 diabetes, elevated Lp(a) levels are associated with an increased risk of cardiovascular events. SUMMARY Although Lp(a) contributes to the development of cardiovascular disease in patients with diabetes, few trials have investigated the benefits of reducing Lp(a) within this patient population. Furthermore, guidelines do not specifically address the risk associated with elevated Lp(a) levels. Despite this, Lp(a) should be measured in patients with diabetes and considered when evaluating their overall risk burden.
Collapse
Affiliation(s)
- Natalie C Ward
- School of Public Health, Curtin University
- School of Medicine, University of Western Australia
| | | | - Gerald F Watts
- School of Medicine, University of Western Australia
- Department of Cardiology, Lipid Disorders Clinic, Royal Perth Hospital, Perth, Australia
| |
Collapse
|
44
|
Wilson DP, Koschinsky ML, Moriarty PM. Expert position statements: comparison of recommendations for the care of adults and youth with elevated lipoprotein(a). Curr Opin Endocrinol Diabetes Obes 2021; 28:159-173. [PMID: 33534258 DOI: 10.1097/med.0000000000000624] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
PURPOSE OF REVIEW Summarize recent recommendations on clinical management of adults and youth with elevated lipoprotein(a) [Lp(a)] who are at-risk of or affected by cardiovascular disease (CVD). RECENT FINDINGS There is ample evidence to support elevated Lp(a) levels, present in approximately 20% of the general population, as a causal, independent risk factor for CVD and its role as a significant risk enhancer. Several guidelines and position statements have been published to assist in the identification, treatment and follow-up of adults with elevated levels of Lp(a). There is growing interest in Lp(a) screening and strategies to improve health behaviors starting in youth, although published recommendations for this population are limited. In addition to the well established increased risk of myocardial infarction, stroke and valvular aortic stenosis, data from the coronavirus pandemic suggest adults with elevated Lp(a) may have a particularly high-risk of cardiovascular complications. Lp(a)-specific-lowering therapies are currently in development. Despite their inability to lower Lp(a), use of statins have been shown to improve outcomes in primary and secondary prevention. SUMMARY Considerable differences exist amongst published guidelines for adults on the use of Lp(a) in clinical practice, and recommendations for youth are limited. With increasing knowledge of Lp(a)'s role in CVD, including recent observations of COVID-19-related risk of cardiovascular complications, more harmonized and comprehensive guidelines for Lp(a) in clinical practice are required. This will facilitate clinical decision-making and help define best practices for identification and management of elevated Lp(a) in adults and youth.
Collapse
Affiliation(s)
- Don P Wilson
- Department of Pediatric Endocrinology, Fort Worth, Texas, USA
| | - Marlys L Koschinsky
- Robarts Research Institute, Schulich School of Medicine and Dentistry, The University of Western Ontario, London, Ontario, Canada
| | - Patrick M Moriarty
- The Department of Internal Medicine, University of Kansas Medical Center, Kansas City, Kansas, USA
| |
Collapse
|
45
|
Anastasiou G, Sakka E, Blathra E, Kalivi A, Elisaf M, Liamis G, Liberopoulos E. Lipoprotein(a): A Concealed Precursor of Increased Cardiovascular Risk? A Real-World Regional Lipid Clinic Experience. Arch Med Res 2021; 52:397-404. [PMID: 33380360 DOI: 10.1016/j.arcmed.2020.12.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Revised: 11/19/2020] [Accepted: 12/03/2020] [Indexed: 12/16/2022]
Abstract
OBJECTIVE Lipoprotein(a) [Lp(a)] is an independent cardiovascular risk factor. We present real-life characteristics of patients with increased Lp(a) levels attending a University Lipid Clinic. METHODS We retrospectively studied patients attending the University of Ioannina Hospital Lipid Clinic with Lp(a) levels ≥30 mg/dL who were followed-up for a median of 22 months. RESULTS One hundred eight patients (median age 59 years, 49% females) were included with median Lp(a) levels 67 mg/dL (30-320). Of patients, 25.1% had established atherosclerotic cardiovascular disease (ASCVD): 11.1 and 5.6% positive personal history of myocardial infarction (MI) and stroke, respectively, 6.5% carotid artery disease and 1.9% lower extremities arterial disease (LEAD). In addition, 35.2% of participants had heterozygous familial hypercholesterolemia (heFH), 37.9% positive family history of premature ASCVD, 29.6% hypertension, 12.0% diabetes and 5.5% chronic kidney disease (CKD). Of patients, 67.6% were receiving statin therapy and 16.6% additional ezetimibe at baseline visit, and 83 and 35% were receiving statin treatment and additional ezetimibe, respectively, during follow-up. Low-density cholesterol (LDL-C) levels and LDL-Ccorrected for Lp(a) levels were significantly reduced in lipid-lowering therapy naive patients by 37 and 40% (p <0.05), in lipid-lowering therapy intensified patients by 31 and 36% (p <0.05), and in patients on stable lipid-lowering treatment by 15% (p <0.05) and 10% (p >0.05), respectively, during follow-up. Lp(a) levels increased by 9% (p <0.05). CONCLUSION Our data confirm the high prevalence of established ASCVD, hFH and positive familial history of premature ASCVD in patients with elevated Lp(a) levels. Lp(a) levels slightly increased during follow-up.
Collapse
Affiliation(s)
- Georgia Anastasiou
- Department of Internal Medicine, University of Ioannina Medical School, Ioannina, Greece
| | - Eftihia Sakka
- Department of Internal Medicine, University of Ioannina Medical School, Ioannina, Greece
| | - Efi Blathra
- Department of Internal Medicine, University of Ioannina Medical School, Ioannina, Greece
| | - Anna Kalivi
- Department of Internal Medicine, University of Ioannina Medical School, Ioannina, Greece
| | - Moses Elisaf
- Department of Internal Medicine, University of Ioannina Medical School, Ioannina, Greece
| | - George Liamis
- Department of Internal Medicine, University of Ioannina Medical School, Ioannina, Greece
| | - Evangelos Liberopoulos
- Department of Internal Medicine, University of Ioannina Medical School, Ioannina, Greece.
| |
Collapse
|
46
|
Lan NSR, Chan DC, Pang J, Fegan PG, Yeap BB, Rankin JM, Schultz CJ, Watts GF, Bell DA. Lipoprotein(a) in Patients With Type 2 Diabetes and Premature Coronary Artery Disease in the Coronary Care Unit. Heart Lung Circ 2020; 30:734-740. [PMID: 33191140 DOI: 10.1016/j.hlc.2020.09.932] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Revised: 08/25/2020] [Accepted: 09/17/2020] [Indexed: 02/03/2023]
Abstract
INTRODUCTION Lipoprotein(a) [Lp(a)] and diabetes are independently associated with premature coronary artery disease (pCAD). However, there is an inverse relationship between Lp(a) concentration and type 2 diabetes (T2D) risk. We examine whether Lp(a) distribution in patients with pCAD differs between those with or without T2D, and whether elevated Lp(a) is associated with pCAD in patients with T2D. METHODS Lp(a) concentration was measured in consecutive acute coronary syndrome (ACS) patients in two coronary care units (study one: ACS with or without diabetes, study two: ACS and diabetes). Elevated Lp(a) mass concentration was defined as ≥0.5 g/L and pCAD where CAD was diagnosed age <60 years. The association between elevated Lp(a) and pCAD was assessed using logistic regression. RESULTS Of 449 patients, 233 (51.9%) had pCAD and 278 (61.9%) had T2D. In patients with pCAD, those with T2D had a significantly lower median Lp(a) concentration (0.13 g/L versus 0.27 g/L, p=0.004). In patients with T2D, elevated Lp(a) was significantly associated with pCAD (OR 2.419, 95% CI 1.513-3.867, p<0.001). After adjusting for gender, smoking, low-density lipoprotein cholesterol, high-density lipoprotein cholesterol and triglycerides, elevated Lp(a) remained significantly associated with pCAD (OR 2.895, 95% CI 1.427-5.876, p=0.003) in patients with T2D. CONCLUSIONS In coronary care patients with pCAD, patients with T2D had lower Lp(a) concentrations than those without T2D. Despite this, elevated Lp(a) remained predictive of pCAD in patients with T2D. Measurement of Lp(a) should be considered in younger adults with T2D to identify who may benefit from earlier preventative therapies to reduce pCAD burden.
Collapse
Affiliation(s)
- Nick S R Lan
- Department of Endocrinology and Diabetes, Fiona Stanley Hospital, Perth, WA, Australia; Department of Cardiology, Fiona Stanley Hospital, Perth, WA, Australia.
| | - Dick C Chan
- Medical School, The University of Western Australia, Perth, WA, Australia
| | - Jing Pang
- Medical School, The University of Western Australia, Perth, WA, Australia
| | - P Gerry Fegan
- Department of Endocrinology and Diabetes, Fiona Stanley Hospital, Perth, WA, Australia; Medical School, Curtin University, Perth, WA, Australia
| | - Bu B Yeap
- Department of Endocrinology and Diabetes, Fiona Stanley Hospital, Perth, WA, Australia; Medical School, The University of Western Australia, Perth, WA, Australia
| | - James M Rankin
- Department of Cardiology, Fiona Stanley Hospital, Perth, WA, Australia
| | - Carl J Schultz
- Medical School, The University of Western Australia, Perth, WA, Australia; Departments of Internal Medicine and Cardiology, Royal Perth Hospital, Perth, WA, Australia
| | - Gerald F Watts
- Medical School, The University of Western Australia, Perth, WA, Australia; Departments of Internal Medicine and Cardiology, Royal Perth Hospital, Perth, WA, Australia
| | - Damon A Bell
- Medical School, The University of Western Australia, Perth, WA, Australia; Departments of Internal Medicine and Cardiology, Royal Perth Hospital, Perth, WA, Australia; Department of Clinical Biochemistry, PathWest Laboratory Medicine WA, Royal Perth and Fiona Stanley Hospitals, Perth, WA, Australia; Department of Biochemistry, Clinipath Pathology, Perth, WA, Australia
| |
Collapse
|
47
|
Katsiki N, Mikhailidis DP. Diabetes and carotid artery disease: a narrative review. ANNALS OF TRANSLATIONAL MEDICINE 2020; 8:1280. [PMID: 33178812 PMCID: PMC7607073 DOI: 10.21037/atm.2019.12.153] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Diabetes mellitus (DM) has been linked to an increased prevalence and severity of carotid artery disease, as well as polyvascular disease. Carotid disease is also associated with obesity and abnormal peri-organ and intra-organ fat (APIFat) deposition (i.e., excess fat accumulation in several organs such as the liver, heart and vessels). In turn, DM is associated with APIFat. The coexistence of these comorbidities confers a greater risk of vascular events. Clinicians should also consider that carotid bruits may predict cardiovascular risk. DM has been related to a greater risk of adverse outcomes after carotid endarterectomy or stenting. Whether modifying risk factors (e.g., glycaemia and dyslipidaemia) in DM patients can improve the outcomes of these procedures needs to be established. Furthermore, DM is a risk factor for contrast-induced acute kidney injury (CI-AKI). The latter should be recorded in DM patients undergoing carotid stenting since it can influence both short- and long-term outcomes. From a pathophysiological perspective, functional changes in the carotid artery may precede morphological ones. Furthermore, carotid plaque characteristics are increasingly being studied in terms of vascular risk stratification and monitoring short-term changes attributed to treatment. The present narrative review discusses the recent (2019) literature on the associations between DM and carotid artery disease. Physicians and vascular surgeons looking after patients with carotid disease and DM should consider these links that may influence outcomes. Further research in this field is also needed to optimise the treatment of such patients.
Collapse
Affiliation(s)
- Niki Katsiki
- Diabetes Center, Division of Endocrinology and Metabolism, First Department of Internal Medicine, AHEPA University Hospital, Medical School Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Dimitri P Mikhailidis
- Department of Clinical Biochemistry, Royal Free Hospital Campus, University College London Medical School, University College London (UCL), London, UK.,Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai, United Arab Emirates
| |
Collapse
|
48
|
Muhanhali D, Zhai T, Cai Z, Ling Y. Lipoprotein(a) concentration is associated with risk of type 2 diabetes and cardiovascular events in a Chinese population with very high cardiovascular risk. Endocrine 2020; 69:63-72. [PMID: 32253681 DOI: 10.1007/s12020-020-02286-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/05/2019] [Accepted: 03/25/2020] [Indexed: 12/26/2022]
Abstract
PURPOSE Evidences have shown that elevated lipoprotein(a) [Lp(a)] levels were associated with a lower risk of type 2 diabetes, but a higher risk of cardiovascular events in general populations. We aim to demonstrate the effect of Lp(a) concentrations on type 2 diabetes and cardiovascular events in a Chinese population with very high cardiovascular risk. METHODS Seven hundred ninety-eight participants who underwent coronary angiography between March and November 2013 with normal glucose metabolism were followed up from July to December 2018. RESULTS Five hundred thirty-five participants completed follow-up, and 395 of them had blood glucose data. Among 395 participants with blood glucose data, a total of 28 incident type 2 diabetes were identified during a median follow-up period of 4.42 years. Compared with the patients in the lowest tertile of Lp(a), the multifactorial adjusted HR for type 2 diabetes was 0.29 in the highest tertile (95% confidence intervals (CI) 0.10-0.89; P for trend = 0.03). Among 535 patients who completed follow-up, a total of 80 cases of major adverse cardiovascular events (MACE) were identified during a median follow-up period of 5.08 years. Compared with the patients in the lowest tertile of Lp(a), the multifactorial adjusted HR for MACE was 1.95 in the highest tertile (95% CI 1.05-3.65; P for trend = 0.03). CONCLUSIONS Elevated Lp(a) levels were associated with a lower risk of type 2 diabetes, but a higher risk of cardiovascular events in a Chinese population with very high cardiovascular risk.
Collapse
Affiliation(s)
- Dilidaer Muhanhali
- Department of Endocrinology and Metabolism, Zhongshan Hospital, Fudan University, No.180 Fenglin Road, Shanghai, 200032, China
| | - Tianyu Zhai
- Department of Endocrinology and Metabolism, Zhongshan Hospital, Fudan University, No.180 Fenglin Road, Shanghai, 200032, China
| | - Zhenqin Cai
- Department of Endocrinology and Metabolism, Zhongshan Hospital, Fudan University, No.180 Fenglin Road, Shanghai, 200032, China
| | - Yan Ling
- Department of Endocrinology and Metabolism, Zhongshan Hospital, Fudan University, No.180 Fenglin Road, Shanghai, 200032, China.
| |
Collapse
|
49
|
Heinrich NS, von Scholten BJ, Reinhard H, Persson F, Ahluwalia TS, Hansen TW, Parving HH, Jacobsen PK, Rossing P. Lipoprotein(a)and renal function decline, cardiovascular disease and mortality in type 2 diabetes and microalbuminuria. J Diabetes Complications 2020; 34:107593. [PMID: 32349898 DOI: 10.1016/j.jdiacomp.2020.107593] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Revised: 03/19/2020] [Accepted: 03/29/2020] [Indexed: 02/06/2023]
Abstract
AIMS Lipoprotein(a)(Lp(a)) has emerged as an independent risk marker for cardiovascular disease (CVD) in the general population and among persons with existing CVD. We investigated associations between serum Lp(a)concentrations and renal function decline, incident CVD and all-cause mortality in individuals with type 2 diabetes (T2D) and microalbuminuria. METHODS Prospective study including 198 individuals with T2D, microalbuminuria and no CVD. Yearly p-creatinine was measured after baseline in 176 of the participants. The renal endpoint was defined as decline in eGFR of >30% from baseline. CVD events and mortality were tracked from national registries. Cox regression analyses were applied both unadjusted and adjusted for traditional risk factors (sex, age, systolic blood pressure, LDL-cholesterol, smoking, HbA1c, creatinine and urinary albumin creatinine ratio (UAER)). RESULTS Baseline mean (SD) age was 59 (9)years, eGFR 89 (17) mL/min/1.73 m2, 77% were male, and median [IQR] UAER was 103 [38-242] mg/24-h. Median Lp(a)was 8.04 [3.42-32.3] mg/dL. Median follow-up was 6.1 years; 38 CVD events, 26 deaths and 43 renal events were recorded. For each doubling of baseline Lp(a), the following hazard ratios (95% confidence intervals) were found before and after adjustment respectively: 0.98 (0.84-1.15) and 1.01 (0.87-1.18) for decline in eGFR > 30%, 0.96 (0.81-1.13) and 0.99 (0.82-1.18) for CVD events, 1.04 (0.85-1.27) and 1.06 (0.87-1.30) for all-cause mortality. CONCLUSIONS In this cohort of individuals with T2D and microalbuminuria, the baseline concentration of Lp(a)was not a risk marker for renal function decline, CVD events or all-cause mortality.
Collapse
Affiliation(s)
| | | | - Henrik Reinhard
- Steno Diabetes Center Copenhagen, Niels Steensensvej 2, 2820 Gentofte, Denmark.
| | - Frederik Persson
- Steno Diabetes Center Copenhagen, Niels Steensensvej 2, 2820 Gentofte, Denmark.
| | | | - Tine Willum Hansen
- Steno Diabetes Center Copenhagen, Niels Steensensvej 2, 2820 Gentofte, Denmark.
| | - Hans-Henrik Parving
- Department of Medical Endocrinology, Rigshospitalet, Copenhagen University Hospital, Blegdamsvej 9, 2100 Copenhagen, Denmark; University of Copenhagen, Copenhagen, Denmark.
| | - Peter Karl Jacobsen
- Department of Cardiology, Rigshospitalet, Copenhagen University Hospital, Blegdamsvej 9, 2100 Copenhagen, Denmark.
| | - Peter Rossing
- Steno Diabetes Center Copenhagen, Niels Steensensvej 2, 2820 Gentofte, Denmark; University of Copenhagen, Copenhagen, Denmark.
| |
Collapse
|
50
|
Lippi G, Favaloro EJ, Sanchis-Gomar F. Antisense lipoprotein[a] therapy: State-of-the-art and future perspectives. Eur J Intern Med 2020; 76:8-13. [PMID: 32336611 DOI: 10.1016/j.ejim.2020.04.036] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2020] [Revised: 04/11/2020] [Accepted: 04/15/2020] [Indexed: 12/11/2022]
Abstract
Several lines of evidence now attest that lipoprotein[a] (Lp[a]) is a significant risk factor for many cardiovascular disorders. This enigmatic lipoprotein, composed of a single copy of apolipoprotein B (apoB) and apolipoprotein[a] (apo [a]), expresses peculiar metabolism, virtually independent from lifestyle interventions. Several therapeutic options have hence been proposed for lowering elevated Lp[a] values, with or without concomitant effect on low density lipoprotein (LDL) particles, mostly encompassing statins, ezetimibe, nicotinic acid, lipoprotein apheresis, and anti-PCSK9 monoclonal antibodies. Since all these medical treatments have some technical and clinical drawbacks, a novel strategy is currently being proposed, based on the use of antisense apo[a] and/or apoB inhibitors. Although the role of these agents in hypercholesterolemic patients is now nearby entering clinical practice, the collection of information on Lp[a] is still underway. Preliminary evidence would suggest that apo[a] antisense therapy seems more appropriate in patients with isolated Lp[a] elevations, while apoB antisense therapy is perhaps more advisable in patients with isolated LDL elevations. In patients with concomitant elevations of Lp[a] and LDL, either combining the two apo[a] and apoB antisense therapies (a strategy which has never been tested), or the combination of well-known and relatively inexpensive drugs such as statins with antisense apo[a] inhibitors can be theoretically suggested. The results of an upcoming phase 3 study with antisense apo[a] inhibitors will hopefully provide definitive clues as to whether this approach may become the standard of care in patients with increased Lp[a] concentrations.
Collapse
Affiliation(s)
- Giuseppe Lippi
- Section of Clinical Biochemistry, University of Verona, Verona, Italy.
| | - Emmanuel J Favaloro
- Department of Haematology, Sydney Centres for Thrombosis and Haemostasis, Institute of Clinical Pathology and Medical Research (ICPMR), NSW Health Pathology, Westmead Hospital, Westmead, New South Wales, Australia
| | - Fabian Sanchis-Gomar
- Department of Physiology, Faculty of Medicine, University of Valencia and INCLIVA Biomedical Research Institute, Valencia, Spain
| |
Collapse
|