1
|
Song CK, Ohlei O, Keller T, Regitz-Zagrosek V, Toepfer S, Steinhagen-Thiessen E, Bertram L, Buchmann N, Demuth I. Lipoprotein(a) and Lung Function Are Associated in Older Adults: Longitudinal and Cross-Sectional Analyses. Biomedicines 2024; 12:1502. [PMID: 39062075 PMCID: PMC11274407 DOI: 10.3390/biomedicines12071502] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Revised: 06/12/2024] [Accepted: 07/01/2024] [Indexed: 07/28/2024] Open
Abstract
While numerous studies have confirmed a causal association between lipoprotein(a) [Lp(a)] and cardiovascular diseases, only a few studies have assessed the relationship between Lp(a) and pulmonary health, with inconsistent findings regarding this topic. This study's aim was to examine whether levels of serum Lp(a) are associated with lung function in a dataset of relatively healthy older adults. We used longitudinal data collected at two time points 7.4 ± 1.5 years apart from 679 participants (52% women, 68 [65-71] years old) from the Berlin Aging Study II (BASE-II). Multiple linear regression models adjusting for covariates were applied to examine the association between Lp(a) and lung function. The forced expiratory volume in one second (FEV1) and the forced vital capacity (FVC) were higher in both men and women with higher Lp(a) levels. However, since this association between lung function parameters and Lp(a) was not supported by Mendelian randomization analyses using recent genome-wide association study data, these relationships should be investigated in future work, as the observed differences are, in part, considerable and potentially clinically relevant.
Collapse
Affiliation(s)
- Chae Kyung Song
- Charité–Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Endocrinology and Metabolic Diseases (Including Division of Lipid Metabolism), Augustenburger Platz 1, 13353 Berlin, Germany
| | - Olena Ohlei
- Lübeck Interdisciplinary Platform for Genome Analytics (LIGA), University of Lübeck, 23562 Lübeck, Germany
| | - Theresa Keller
- Institute of Biometry and Clinical Epidemiology, Charité–Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Reinhardtstraße 58, 10117 Berlin, Germany
| | - Vera Regitz-Zagrosek
- Institute for Gender in Medicine, Center for Cardiovascular Research, Charité–Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, 10099 Berlin, Germany
- Department of Cardiology, University Hospital Zürich, University of Zürich, 8057 Zürich, Switzerland
| | - Sarah Toepfer
- Charité–Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Endocrinology and Metabolic Diseases (Including Division of Lipid Metabolism), Augustenburger Platz 1, 13353 Berlin, Germany
| | - Elisabeth Steinhagen-Thiessen
- Charité–Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Endocrinology and Metabolic Diseases (Including Division of Lipid Metabolism), Augustenburger Platz 1, 13353 Berlin, Germany
| | - Lars Bertram
- Lübeck Interdisciplinary Platform for Genome Analytics (LIGA), University of Lübeck, 23562 Lübeck, Germany
| | - Nikolaus Buchmann
- Department of Cardiology, Charité–University Medicine Berlin, Campus Benjamin Franklin, 10117 Berlin, Germany
| | - Ilja Demuth
- Charité–Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Endocrinology and Metabolic Diseases (Including Division of Lipid Metabolism), Augustenburger Platz 1, 13353 Berlin, Germany
- Charité–Universitätsmedizin Berlin, BCRT-Berlin Institute of Health Center for Regenerative Therapies, 10117 Berlin, Germany
| |
Collapse
|
2
|
Groenen AG, Matveyenko A, Matienzo N, Halmos B, Zhang H, Westerterp M, Reyes-Soffer G. Apolipoprotein(a) production and clearance are associated with plasma IL-6 and IL-18 levels, dependent on ethnicity. Atherosclerosis 2024; 391:117474. [PMID: 38428286 DOI: 10.1016/j.atherosclerosis.2024.117474] [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: 09/19/2023] [Revised: 01/31/2024] [Accepted: 02/01/2024] [Indexed: 03/03/2024]
Abstract
BACKGROUND AND AIMS High plasma lipoprotein (a) [Lp(a)] levels are associated with increased atherosclerotic cardiovascular disease (ASCVD), in part attributed to elevated inflammation. High plasma Lp(a) levels inversely correlate with apolipoprotein (a) [(APO(a)] isoform size. APO(a) isoform size is negatively associated with APO(a) production rate (PR) and positively associated with APO(a) fractional catabolic rate (FCR). We asked whether APO(a) PR and FCR (kinetics) are associated with plasma levels of interleukin (IL)-6 and IL-18, pro-inflammatory interleukins that promote ASCVD. METHODS We used samples from existing data of APO(a) kinetic studies from an ethnically diverse cohort (n = 25: 10 Black, 9 Hispanic, and 6 White subjects) and assessed IL-6 and IL-18 plasma levels. We performed multivariate linear regression analyses to examine the relationships between predictors APO(a) PR or APO(a) FCR, and outcome variables IL-6 or IL-18. In these analyses, we adjusted for parameters known to affect Lp(a) levels and APO(a) PR and FCR, including race/ethnicity and APO(a) isoform size. RESULTS APO(a) PR and FCR were positively associated with plasma IL-6, independent of isoform size, and dependent on race/ethnicity. APO(a) PR was positively associated with plasma IL-18, independent of isoform size and race/ethnicity. APO(a) FCR was not associated with plasma IL-18. CONCLUSIONS Our studies demonstrate a relationship between APO(a) PR and FCR and plasma IL-6 or IL-18, interleukins that promote ASCVD. These studies provide new insights into Lp(a) pro-inflammatory properties and are especially relevant in view of therapies targeting APO(a) to decrease cardiovascular risk.
Collapse
Affiliation(s)
- Anouk G Groenen
- Department of Pediatrics, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Anastasiya Matveyenko
- Columbia University Irving Medical Center, College of Physicians and Surgeons, Department of Medicine, Division of Preventive Medicine and Nutrition, New York, NY, USA
| | - Nelsa Matienzo
- Columbia University Irving Medical Center, College of Physicians and Surgeons, Department of Medicine, Division of Preventive Medicine and Nutrition, New York, NY, USA
| | - Benedek Halmos
- Department of Pediatrics, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Hanrui Zhang
- Columbia University Irving Medical Center, Division of Cardiology, New York, NY, USA
| | - Marit Westerterp
- Department of Pediatrics, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands.
| | - Gissette Reyes-Soffer
- Columbia University Irving Medical Center, College of Physicians and Surgeons, Department of Medicine, Division of Preventive Medicine and Nutrition, New York, NY, USA.
| |
Collapse
|
3
|
Fogacci F, Di Micoli V, Sabouret P, Giovannini M, Cicero AFG. Lifestyle and Lipoprotein(a) Levels: Does a Specific Counseling Make Sense? J Clin Med 2024; 13:751. [PMID: 38337445 PMCID: PMC10856708 DOI: 10.3390/jcm13030751] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Revised: 01/20/2024] [Accepted: 01/26/2024] [Indexed: 02/12/2024] Open
Abstract
Lipoprotein(Lp)(a) is a variant of low-density lipoprotein (LDL), bound to apolipoprotein B100, whose levels are associated with a significant increase in the risk of atherosclerosis-related cardiovascular events, but also to aortic stenosis and atrial fibrillation. Since plasma levels of Lp(a) are commonly considered resistant to lifestyle changes, we critically reviewed the available evidence on the effect of weight loss, dietary supplements, and physical activity on this risk factor. In our review, we observed that relevant body weight loss, a relatively high intake of saturated fatty acids, the consumption of red wine, and intense physical exercise seems to be associated with significantly lower plasma Lp(a) levels. On the contrary, foods rich in trans-unsaturated fatty acids are associated with increased Lp(a) levels. With regard to dietary supplements, coenzyme Q10, L-Carnitine, and flaxseed exert a mild but significant lowering effect on plasma Lp(a).
Collapse
Affiliation(s)
- Federica Fogacci
- Hypertension and Cardiovascular Risk Research Center, Medical and Surgical Sciences Department, Alma Mater Studiorum University of Bologna, 40100 Bologna, Italy; (V.D.M.); (M.G.)
| | - Valentina Di Micoli
- Hypertension and Cardiovascular Risk Research Center, Medical and Surgical Sciences Department, Alma Mater Studiorum University of Bologna, 40100 Bologna, Italy; (V.D.M.); (M.G.)
| | - Pierre Sabouret
- INSERM UMRS_1166, Cardiology Institute, Pitié Salpêtrière Hospital (AP-HP), ACTION Study Group, Sorbonne University, 75013 Paris, France;
| | - Marina Giovannini
- Hypertension and Cardiovascular Risk Research Center, Medical and Surgical Sciences Department, Alma Mater Studiorum University of Bologna, 40100 Bologna, Italy; (V.D.M.); (M.G.)
| | - Arrigo F. G. Cicero
- Hypertension and Cardiovascular Risk Research Center, Medical and Surgical Sciences Department, Alma Mater Studiorum University of Bologna, 40100 Bologna, Italy; (V.D.M.); (M.G.)
- Cardiovascular Medicine Unit, Heart, Thoracic and Vascular Department, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40100 Bologna, Italy
| |
Collapse
|
4
|
Liang D, Liang D, Liu J, Zheng Y, Huang D, Li Z, Huang X, Chen J. Impact of lipoprotein(a) and fibrinogen on prognosis in patients with coronary artery disease: A retrospective cohort study. Technol Health Care 2024; 32:3317-3328. [PMID: 38848204 PMCID: PMC11492067 DOI: 10.3233/thc-240005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Accepted: 04/16/2024] [Indexed: 06/09/2024]
Abstract
BACKGROUND Despite the considerable progress made in preventative methods, medication, and interventional therapies, it remains evident that cardiovascular events (CVEs) continue to be the primary cause of both death and morbidity among individuals diagnosed with coronary artery disease (CAD). OBJECTIVE To compare the connection between lipoprotein a (Lp[a]), fibrinogen (Fib), and both parameters combined with all-cause mortality to detect their value as prognostic biomarkers. METHODS This is a retrospective study. Patients diagnosed with CAD between January 2007 and December 2020 at the Guangdong Provincial People's Hospital (China) were involved in the study. 43,367 patients met the eligibility criteria. The Lp(a) and Fib levels were distributed into three tertile groups (low, medium, and high). All of the patients included in the study were followed up for all-cause mortality. Kaplan-Meier and Cox regression were performed to determine the relationship between Lp(a), Fib, and all-cause mortality. A concordance statistics model was developed to detect the impact of Fib and Lp(a) in terms of anticipating poor outcomes in patients with CAD. RESULTS Throughout a median follow-up of 67.0 months, 6,883 (15.9%) patients died. Participants with high Lp(a) (above 27.60 mg/dL) levels had a significantly higher risk for all-cause mortality than individuals with low Lp(a) levels (below 11.13 mg/dL; adjusted hazard ratio [aHR] 1.219, 95% confidence interval [CI]: 1.141-1.304, p< 0.001). Similarly, patients with high Fib levels (above 4.32 g/L) had a significantly greater risk of developing all-cause mortality compared with those with reduced Fib levels (below 3.41 g/L; aHR 1.415, 95% CI: 1.323-1.514, p< 0.001). Patients with raised Lp(a) and Fib levels had the maximum risk for all-cause mortality (aHR 1.702; 95% CI: 1.558-1.859, p< 0.001). When considered together, Lp(a) and Fib caused a significant elevation of the concordance statistic by 0.009 (p< 0.05), suggesting a higher value for predicting mortality when combining the two indicators. CONCLUSION High Lp(a) and Fib levels could be used as predictive biomarkers for all-cause mortality in individuals with CAD. The prediction accuracy for all-cause mortality improved after combining the two parameters.
Collapse
Affiliation(s)
- Dakai Liang
- Department of Cardiology, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
- Department of Cardiology, People’s Hospital of Yangjiang, Yangjiang, China
| | - Dandan Liang
- Department of Cardiology, People’s Hospital of Yangjiang, Yangjiang, China
| | - Jin Liu
- Department of Cardiology, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Yiying Zheng
- Department of Cardiology, People’s Hospital of Yangjiang, Yangjiang, China
| | - Dehua Huang
- Department of Cardiology, People’s Hospital of Yangjiang, Yangjiang, China
| | - Zeliang Li
- Department of Cardiology, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Xiaoyu Huang
- Department of Cardiology, People’s Hospital of Yangjiang, Yangjiang, China
| | - Jiyan Chen
- Department of Cardiology, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| |
Collapse
|
5
|
Cicero AFG, Fogacci F, Giovannini M, Grandi E, D’Addato S, Borghi C. Estimating the Prevalence and Characteristics of Patients Potentially Eligible for Lipoprotein(a)-Lowering Therapies in a Real-World Setting. Biomedicines 2023; 11:3289. [PMID: 38137510 PMCID: PMC10741849 DOI: 10.3390/biomedicines11123289] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Revised: 12/10/2023] [Accepted: 12/11/2023] [Indexed: 12/24/2023] Open
Abstract
High lipoprotein(a) (Lp(a)) plasma levels are significantly associated with an increased risk of developing atherosclerotic cardiovascular diseases (ASCVD). The aim of this analysis was to estimate the prevalence and characteristics of patients potentially eligible for Lp(a)-lowering therapies in a real-world setting (i.e., patients with ASCVD and Lp(a) levels > 70 mg/dL). For this reason, we pooled data from a large cohort of Italian outpatients (N = 5961; men: 2879, women: 3982) with dyslipidemia. A binary logistic regression analysis was used to determine the significant predictors of ASCVD in the cohort, which were age (Odds Ratio (OR): 1.158, 95% Confidence Interval (CI): 1.114 to 1.203, p < 0.001), low-density lipoprotein cholesterol at entry (OR: 1.989, 95% CI: 1.080 to 1.198, p = 0.020) and Lp(a) (OR: 1.090, 95% CI: 1.074 to 1.107, p < 0.001). In our cohort, almost half of patients with ASCVD (44.7%) may be eligible to be treated with Lp(a)-lowering agents. Interestingly, patients who do not meet the treatment criteria despite high Lp(a) (50-70 mg/dL), respectively, account for 4.7% and 7.3% of those in primary and secondary ASCVD prevention. In conclusion, in our large cohort of outpatients with dyslipidemia, the prevalence of individuals with ASCVD and very high Lp(a) plasma levels is quite high, even with a conservative estimation.
Collapse
Affiliation(s)
- Arrigo F. G. Cicero
- Hypertension and Cardiovascular Risk Factors Research Centre, Medical and Surgical Sciences Department, Alma Mater Studiorum University of Bologna, 40100 Bologna, Italy; (A.F.G.C.); (M.G.); (E.G.); (S.D.); (C.B.)
- Cardiovascular Medicine Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy
| | - Federica Fogacci
- Hypertension and Cardiovascular Risk Factors Research Centre, Medical and Surgical Sciences Department, Alma Mater Studiorum University of Bologna, 40100 Bologna, Italy; (A.F.G.C.); (M.G.); (E.G.); (S.D.); (C.B.)
- Cardiovascular Medicine Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy
| | - Marina Giovannini
- Hypertension and Cardiovascular Risk Factors Research Centre, Medical and Surgical Sciences Department, Alma Mater Studiorum University of Bologna, 40100 Bologna, Italy; (A.F.G.C.); (M.G.); (E.G.); (S.D.); (C.B.)
| | - Elisa Grandi
- Hypertension and Cardiovascular Risk Factors Research Centre, Medical and Surgical Sciences Department, Alma Mater Studiorum University of Bologna, 40100 Bologna, Italy; (A.F.G.C.); (M.G.); (E.G.); (S.D.); (C.B.)
- Cardiovascular Medicine Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy
| | - Sergio D’Addato
- Hypertension and Cardiovascular Risk Factors Research Centre, Medical and Surgical Sciences Department, Alma Mater Studiorum University of Bologna, 40100 Bologna, Italy; (A.F.G.C.); (M.G.); (E.G.); (S.D.); (C.B.)
- Cardiovascular Medicine Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy
| | - Claudio Borghi
- Hypertension and Cardiovascular Risk Factors Research Centre, Medical and Surgical Sciences Department, Alma Mater Studiorum University of Bologna, 40100 Bologna, Italy; (A.F.G.C.); (M.G.); (E.G.); (S.D.); (C.B.)
- Cardiovascular Medicine Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy
| |
Collapse
|
6
|
Fogacci F, Di Micoli V, Avagimyan A, Giovannini M, Imbalzano E, Cicero AFG. Assessment of Apolipoprotein(a) Isoform Size Using Phenotypic and Genotypic Methods. Int J Mol Sci 2023; 24:13886. [PMID: 37762189 PMCID: PMC10531419 DOI: 10.3390/ijms241813886] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Revised: 09/03/2023] [Accepted: 09/06/2023] [Indexed: 09/29/2023] Open
Abstract
Apolipoprotein(a) (apo(a)) is the protein component that defines lipoprotein(a) (Lp(a)) particles and is encoded by the LPA gene. The apo(a) is extremely heterogeneous in size due to the copy number variations in the kringle-IV type 2 (KIV2) domains. In this review, we aim to discuss the role of genetics in establishing Lp(a) as a risk factor for coronary heart disease (CHD) by examining a series of molecular biology techniques aimed at identifying the best strategy for a possible application in clinical research and practice, according to the current gold standard.
Collapse
Affiliation(s)
- Federica Fogacci
- Hypertension and Cardiovascular Risk Research Center, Medical and Surgical Sciences Department, Alma Mater Studiorum University of Bologna, 40138 Bologna, Italy; (F.F.); (V.D.M.); (M.G.)
| | - Valentina Di Micoli
- Hypertension and Cardiovascular Risk Research Center, Medical and Surgical Sciences Department, Alma Mater Studiorum University of Bologna, 40138 Bologna, Italy; (F.F.); (V.D.M.); (M.G.)
| | - Ashot Avagimyan
- Pathological Anatomy Department, Yerevan State Medical University, Yerevan 0025, Armenia;
| | - Marina Giovannini
- Hypertension and Cardiovascular Risk Research Center, Medical and Surgical Sciences Department, Alma Mater Studiorum University of Bologna, 40138 Bologna, Italy; (F.F.); (V.D.M.); (M.G.)
| | - Egidio Imbalzano
- Department of Clinical and Experimental Medicine, University of Messina, 98125 Messina, Italy;
| | - Arrigo F. G. Cicero
- Hypertension and Cardiovascular Risk Research Center, Medical and Surgical Sciences Department, Alma Mater Studiorum University of Bologna, 40138 Bologna, Italy; (F.F.); (V.D.M.); (M.G.)
- Cardiovascular Medicine Unit, Heart, Thoracic and Vascular Department, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40100 Bologna, Italy
| |
Collapse
|
7
|
Al Hageh C, Chacar S, Ghassibe-Sabbagh M, Platt DE, Henschel A, Hamdan H, Gauguier D, El Murr Y, Alefishat E, Chammas E, O’Sullivan S, Abchee A, Nader M, Zalloua PA. Elevated Lp(a) Levels Correlate with Severe and Multiple Coronary Artery Stenotic Lesions. Vasc Health Risk Manag 2023; 19:31-41. [PMID: 36703868 PMCID: PMC9871050 DOI: 10.2147/vhrm.s394134] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Accepted: 12/21/2022] [Indexed: 01/19/2023] Open
Abstract
Backgrounds and Aims The role of Lipoprotein(a) (Lp(a)) in increasing the risk of cardiovascular diseases is reported in several populations. The aim of this study is to investigate the correlation of high Lp(a) levels with the degree of coronary artery stenosis. Methods Two hundred and sixty-eight patients were enrolled for this study. Patients who underwent coronary artery angiography and who had Lp(a) measurements available were included in this study. Binomial logistic regressions were applied to investigate the association between Lp(a) and stenosis in the four major coronary arteries. The effect of LDL and HDL Cholesterol on modulating the association of Lp(a) with coronary artery disease (CAD) was also evaluated. Multinomial regression analysis was applied to assess the association of Lp(a) with the different degrees of stenosis in the four major coronary arteries. Results Our analyses showed that Lp(a) is a risk factor for CAD and this risk is significantly apparent in patients with HDL-cholesterol ≥35 mg/dL and in non-obese patients. A large proportion of the study patients with elevated Lp(a) levels had CAD even when exhibiting high HDL serum levels. Increased HDL with low Lp(a) serum levels were the least correlated with stenosis. A significantly higher levels of Lp(a) were found in patients with >50% stenosis in at least two major coronary vessels arguing for pronounced and multiple stenotic lesions. Finally, the derived variant (rs1084651) of the LPA gene was significantly associated with CAD. Conclusion Our study highlights the importance of Lp(a) levels as an independent biological marker of severe and multiple coronary artery stenosis.
Collapse
Affiliation(s)
- Cynthia Al Hageh
- Department of Molecular Biology and Genetics, College of Medicine and Health Sciences, Khalifa University of Science and Technology, Abu Dhabi, United Arab Emirates
| | - Stephanie Chacar
- Department of Physiology and Immunology, College of Medicine and Health Sciences, Khalifa University of Science and Technology, Abu Dhabi, United Arab Emirates
| | - Michella Ghassibe-Sabbagh
- Department of Natural Sciences, School of Arts and Sciences, Lebanese American University, Beirut, Lebanon
| | - Daniel E Platt
- Computational Biology Center, IBM TJ Watson Research Centre, Yorktown Hgts, NY, USA
| | - Andreas Henschel
- Department of Electrical Engineering and Computer, Khalifa University of Science and Technology, Abu Dhabi, United Arab Emirates
| | - Hamdan Hamdan
- Department of Physiology and Immunology, College of Medicine and Health Sciences, Khalifa University of Science and Technology, Abu Dhabi, United Arab Emirates
| | - Dominique Gauguier
- Université Paris Cité, INSERM UMR 1124, Paris, 75006, France,McGill University and Genome Quebec Innovation Centre, Montreal, QC, H3A 0G1, Canada
| | - Yara El Murr
- Department of Natural Sciences, School of Arts and Sciences, Lebanese American University, Beirut, Lebanon
| | - Eman Alefishat
- Department of Pharmacology, College of Medicine and Health Sciences, Khalifa University of Science and Technology, Abu Dhabi, United Arab Emirates,Center for Biotechnology, Khalifa University of Science and Technology, Abu Dhabi, 127788, United Arab Emirates
| | - Elie Chammas
- School of Medicine, Lebanese University, Beirut, Lebanon
| | - Siobhán O’Sullivan
- Department of Molecular Biology and Genetics, College of Medicine and Health Sciences, Khalifa University of Science and Technology, Abu Dhabi, United Arab Emirates
| | - Antoine Abchee
- Sheikh Shakhbout Medical City, Abu Dhabi, United Arab Emirates
| | - Moni Nader
- Department of Physiology and Immunology, College of Medicine and Health Sciences, Khalifa University of Science and Technology, Abu Dhabi, United Arab Emirates,Center for Biotechnology, Khalifa University of Science and Technology, Abu Dhabi, 127788, United Arab Emirates
| | - Pierre A Zalloua
- Department of Molecular Biology and Genetics, College of Medicine and Health Sciences, Khalifa University of Science and Technology, Abu Dhabi, United Arab Emirates,Center for Biotechnology, Khalifa University of Science and Technology, Abu Dhabi, 127788, United Arab Emirates,Harvard T.H. Chan School of Public Health, Boston, MA, USA,Correspondence: Pierre A Zalloua; Moni Nader, College of Medicine and Health Sciences, Khalifa University for Science and Technology, PO Box 127788, Abu Dhabi, United Arab Emirates, Email ;
| |
Collapse
|
8
|
Harrington RA. Trials and Tribulations of Randomized Clinical Trials. Circulation 2022; 146:380-382. [PMID: 35775415 DOI: 10.1161/circulationaha.122.060649] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
9
|
Phua K, Chew NWS, Kong WKF, Tan RS, Ye L, Poh KK. The mechanistic pathways of oxidative stress in aortic stenosis and clinical implications. Theranostics 2022; 12:5189-5203. [PMID: 35836811 PMCID: PMC9274751 DOI: 10.7150/thno.71813] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Accepted: 06/03/2022] [Indexed: 02/06/2023] Open
Abstract
Despite the elucidation of the pathways behind the development of aortic stenosis (AS), there remains no effective medical treatment to slow or reverse its progress. Instead, the gold standard of care in severe or symptomatic AS is replacement of the aortic valve. Oxidative stress is implicated, both directly as well as indirectly, in lipid infiltration, inflammation and fibro-calcification, all of which are key processes underlying the pathophysiology of degenerative AS. This culminates in the breakdown of the extracellular matrix, differentiation of the valvular interstitial cells into an osteogenic phenotype, and finally, calcium deposition as well as thickening of the aortic valve. Oxidative stress is thus a promising and potential therapeutic target for the treatment of AS. Several studies focusing on the mitigation of oxidative stress in the context of AS have shown some success in animal and in vitro models, however similar benefits have yet to be seen in clinical trials. Statin therapy, once thought to be the key to the treatment of AS, has yielded disappointing results, however newer lipid lowering therapies may hold some promise. Other potential therapies, such as manipulation of microRNAs, blockade of the renin-angiotensin-aldosterone system and the use of dipeptidylpeptidase-4 inhibitors will also be reviewed.
Collapse
Affiliation(s)
- Kailun Phua
- Department of Cardiology, National University Heart Centre, National University Hospital, Singapore, Singapore
| | - Nicholas WS Chew
- Department of Cardiology, National University Heart Centre, National University Hospital, Singapore, Singapore,✉ Corresponding authors: A/Prof Kian-Keong Poh, . Dr Nicholas Chew, MBChB, MMED (Singapore), MRCP (UK) . Department of Cardiology, National University Heart Centre Singapore, National University Health System, Singapore. 1E Kent Ridge Rd, NUHS Tower Block, Level 9, Singapore 119228. Fax: (65) 68722998 Telephone: (65) 67722476
| | - William KF Kong
- Department of Cardiology, National University Heart Centre, National University Hospital, Singapore, Singapore
| | - Ru-San Tan
- Department of Cardiology, National Heart Centre Singapore, Singapore, 169609, Singapore
| | - Lei Ye
- National Heart Research Institute Singapore, National Heart Centre Singapore, Singapore, 169609, Singapore
| | - Kian-Keong Poh
- Department of Cardiology, National University Heart Centre, National University Hospital, Singapore, Singapore,Yong Loo Lin School of Medicine, National University of Singapore, Singapore,✉ Corresponding authors: A/Prof Kian-Keong Poh, . Dr Nicholas Chew, MBChB, MMED (Singapore), MRCP (UK) . Department of Cardiology, National University Heart Centre Singapore, National University Health System, Singapore. 1E Kent Ridge Rd, NUHS Tower Block, Level 9, Singapore 119228. Fax: (65) 68722998 Telephone: (65) 67722476
| |
Collapse
|
10
|
Tsaban G. Statins and lipoprotein(a); facing the residual risk. Eur J Prev Cardiol 2021; 29:777-778. [PMID: 34904162 DOI: 10.1093/eurjpc/zwab178] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Affiliation(s)
- Gal Tsaban
- Department of Public Health, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beersheba, Israel.,Department of Cardiology, Heart Institute, Rager Av. 101, Beersheba, 84101, Israel
| |
Collapse
|
11
|
Banach M, Burchardt P, Chlebus K, Dobrowolski P, Dudek D, Dyrbuś K, Gąsior M, Jankowski P, Jóźwiak J, Kłosiewicz-Latoszek L, Kowalska I, Małecki M, Prejbisz A, Rakowski M, Rysz J, Solnica B, Sitkiewicz D, Sygitowicz G, Sypniewska G, Tomasik T, Windak A, Zozulińska-Ziółkiewicz D, Cybulska B. PoLA/CFPiP/PCS/PSLD/PSD/PSH guidelines on diagnosis and therapy of lipid disorders in Poland 2021. Arch Med Sci 2021; 17:1447-1547. [PMID: 34900032 PMCID: PMC8641518 DOI: 10.5114/aoms/141941] [Citation(s) in RCA: 88] [Impact Index Per Article: 29.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Accepted: 11/03/2021] [Indexed: 12/18/2022] Open
Abstract
In Poland there are still nearly 20 million individuals with hypercholesterolaemia, most of them are unaware of their condition; that is also why only ca. 5% of patients with familial hypercholesterolaemia have been diagnosed; that is why other rare cholesterol metabolism disorders are so rarely diagnosed in Poland. Let us hope that these guidelines, being an effect of work of experts representing 6 main scientific societies, as well as the network of PoLA lipid centers being a part of the EAS lipid centers, certification of lipidologists by PoLA, or the growing number of centers for rare diseases, with a network planned by the Ministry of Health, improvements in coordinated care for patients after myocardial infarction (KOS-Zawał), reimbursement of innovative agents, as well as introduction in Poland of an effective primary prevention program, will make improvement in relation to these unmet needs in diagnostics and treatment of lipid disorders possible.
Collapse
Affiliation(s)
- Maciej Banach
- Department of Preventive Cardiology and Lipidology, Medical University of Lodz, Lodz, Poland
- Cardiovascular Research Center, University of Zielona Gora, Zielona Gora, Poland
- Department of Cardiology and Congenital Diseases of Adults, Polish Mother’s Memorial Hospital Research Institute (PMMHRI) in Lodz, Lodz, Poland
| | - Paweł Burchardt
- Department of Hypertensiology, Angiology, and Internal Medicine, K. Marcinkowski Poznan University of Medical Science, Poznan, Poland
- Department of Cardiology, Cardiovascular Unit, J. Strus Hospital, Poznan, Poland
| | - Krzysztof Chlebus
- First Department and Chair of Cardiology, Medical University of Gdansk, Gdansk, Poland
| | - Piotr Dobrowolski
- Department of Hypertension, National Institute of Cardiology, Warsaw, Poland
| | - Dariusz Dudek
- Institute of Cardiology, Jagiellonian University Medical College, Krakow, Poland
| | - Krzysztof Dyrbuś
- 3 Department of Cardiology, Faculty of Medical Sciences in Zabrze, Medical University of Silesia, Katowice, Poland; Silesian Center for Heart Diseases in Zabrze, Poland
| | - Mariusz Gąsior
- 3 Department of Cardiology, Faculty of Medical Sciences in Zabrze, Medical University of Silesia, Katowice, Poland; Silesian Center for Heart Diseases in Zabrze, Poland
| | - Piotr Jankowski
- Department of Internal Medicine and Geriatric Cardiology, Centre of Postgraduate Medical Education, Warsaw, Poland
- Department of Cardiology and Arterial Hypertension, Institute of Cardiology, Jagiellonian University Medical College, Krakow, Poland
| | - Jacek Jóźwiak
- Department of Family Medicine and Public Health, Institute of Medical Sciences, Faculty of Medicine, University of Opole, Opole, Poland
| | | | - Irina Kowalska
- Department of Internal Medicine and Metabolic Diseases, Medical University of Bialystok, Bialystok, Poland
| | - Maciej Małecki
- Department and Chair of Metabolic Diseases, Jagiellonian University Medical College, Krakow, Poland
| | - Aleksander Prejbisz
- Department of Hypertension, National Institute of Cardiology, Warsaw, Poland
| | - Michał Rakowski
- Department of Molecular Biophysics, Institute of Biophysics, Faculty of Biology and Environmental Protection, University of Lodz, Lodz, Poland
| | - Jacek Rysz
- Chair of Nephrology, Arterial Hypertension, and Family Medicine, Medical University of Lodz, Lodz, Poland
| | - Bogdan Solnica
- Chair of Clinical Biochemistry, Jagiellonian University Medical College, Krakow, Poland
| | - Dariusz Sitkiewicz
- Department of Clinical Chemistry and Laboratory Diagnostics, Medical University of Warsaw, Warsaw, Poland
| | - Grażyna Sygitowicz
- Department of Clinical Chemistry and Laboratory Diagnostics, Medical University of Warsaw, Warsaw, Poland
| | - Grażyna Sypniewska
- Department of Laboratory Medicine, L. Rydygier Medical College in Bydgoszcz, Nicolaus Copernicus University in Torun, Poland
| | - Tomasz Tomasik
- Chair of Family Medicine, Jagiellonian University Medical College, Krakow, Poland
| | - Adam Windak
- Chair of Family Medicine, Jagiellonian University Medical College, Krakow, Poland
| | - Dorota Zozulińska-Ziółkiewicz
- Department and Chair of Internal Medicine and Diabetology, K. Marcinkowski Poznan University of Medical Sciences, Poznan, Poland
| | - Barbara Cybulska
- National Institute of Public Health NIH – National Research Institute, Warsaw, Poland
| |
Collapse
|
12
|
van Zuydam NR, Stiby A, Abdalla M, Austin E, Dahlström EH, McLachlan S, Vlachopoulou E, Ahlqvist E, Di Liao C, Sandholm N, Forsblom C, Mahajan A, Robertson NR, Rayner NW, Lindholm E, Sinisalo J, Perola M, Kallio M, Weiss E, Price J, Paterson A, Klein B, Salomaa V, Palmer CN, Groop PH, Groop L, McCarthy MI, de Andrade M, Morris AP, Hopewell JC, Colhoun HM, Kullo IJ. Genome-Wide Association Study of Peripheral Artery Disease. CIRCULATION. GENOMIC AND PRECISION MEDICINE 2021; 14:e002862. [PMID: 34601942 PMCID: PMC8542067 DOI: 10.1161/circgen.119.002862] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Accepted: 08/31/2021] [Indexed: 12/24/2022]
Abstract
BACKGROUND Peripheral artery disease (PAD) affects >200 million people worldwide and is associated with high mortality and morbidity. We sought to identify genomic variants associated with PAD overall and in the contexts of diabetes and smoking status. METHODS We identified genetic variants associated with PAD and then meta-analyzed with published summary statistics from the Million Veterans Program and UK Biobank to replicate their findings. Next, we ran stratified genome-wide association analysis in ever smokers, never smokers, individuals with diabetes, and individuals with no history of diabetes and corresponding interaction analyses, to identify variants that modify the risk of PAD by diabetic or smoking status. RESULTS We identified 5 genome-wide significant (Passociation ≤5×10-8) associations with PAD in 449 548 (Ncases=12 086) individuals of European ancestry near LPA (lipoprotein [a]), CDKN2BAS1 (CDKN2B antisense RNA 1), SH2B3 (SH2B adaptor protein 3) - PTPN11 (protein tyrosine phosphatase non-receptor type 11), HDAC9 (histone deacetylase 9), and CHRNA3 (cholinergic receptor nicotinic alpha 3 subunit) loci (which overlapped previously reported associations). Meta-analysis with variants previously associated with PAD showed that 18 of 19 published variants remained genome-wide significant. In individuals with diabetes, rs116405693 at the CCSER1 (coiled-coil serine rich protein 1) locus was associated with PAD (odds ratio [95% CI], 1.51 [1.32-1.74], Pdiabetes=2.5×10-9, Pinteractionwithdiabetes=5.3×10-7). Furthermore, in smokers, rs12910984 at the CHRNA3 locus was associated with PAD (odds ratio [95% CI], 1.15 [1.11-1.19], Psmokers=9.3×10-10, Pinteractionwithsmoking=3.9×10-5). CONCLUSIONS Our analyses confirm the published genetic associations with PAD and identify novel variants that may influence susceptibility to PAD in the context of diabetes or smoking status.
Collapse
Affiliation(s)
- Natalie R. van Zuydam
- Department of Immunology, Genetics and Pathology, Science for Life Laboratory, Uppsala University, Sweden (N.R.v.Z.)
- Wellcome Centre for Human Genetics, Nuffield Department of Medicine (N.R.v.Z., M.A., A.M., N.R.R., N.W.R., M.I.M., A.P.M.), University of Oxford, United Kingdom
- Oxford Centre for Diabetes, Endocrinology and Metabolism, Radcliffe Department of Medicine (N.R.v.Z., A.M., N.R.R., N.W.R., M.I.M.), University of Oxford, United Kingdom
| | - Alexander Stiby
- Clinical Trial Service Unit and Epidemiological Studies Unit, Nuffield Department of Population Health (A.S., J.C.H.), University of Oxford, United Kingdom
| | - Moustafa Abdalla
- Wellcome Centre for Human Genetics, Nuffield Department of Medicine (N.R.v.Z., M.A., A.M., N.R.R., N.W.R., M.I.M., A.P.M.), University of Oxford, United Kingdom
| | - Erin Austin
- Department of Cardiovascular Medicine and the Gonda Vascular Center, Mayo Clinic, Rochester, MN (E. Austin, M.d.A., I.J.K.)
| | - Emma H. Dahlström
- Folkhälsan Institute of Genetics, Folkhälsan Research Center, Helsinki, Finland (E.H.D., N.S., C.F., P.-H.G.)
- Abdominal Center, Nephrology (E.H.D., N.S., C.F., P.-H.G.), University of Helsinki, Finland
- Helsinki University Hospital, Research Program for Clinical and Molecular Metabolism, Faculty of Medicine (E.H.D., N.S., C.F., P.-H.G.), University of Helsinki, Finland
| | - Stela McLachlan
- Usher Institute of Population Health Sciences and Informatics, The University of Edinburgh, United Kingdom (S.M., E.W., J.P.)
| | - Efthymia Vlachopoulou
- Department of Medicine, Helsinki University Central Hospital (E.V.), University of Helsinki, Finland
| | - Emma Ahlqvist
- Genomics, Diabetes and Endocrinology, Lund University Diabetes Centre, Malmö, Sweden (E. Ahlqvist, E.L., L.G.)
| | - Chen Di Liao
- Dalla Lana School of Public Health, University of Toronto, ON, Canada (C.D.L., A.P.)
- Genetics & Genome Biology, SickKids, Toronto, ON, Canada (C.D.L., A.P.)
| | - Niina Sandholm
- Folkhälsan Institute of Genetics, Folkhälsan Research Center, Helsinki, Finland (E.H.D., N.S., C.F., P.-H.G.)
- Abdominal Center, Nephrology (E.H.D., N.S., C.F., P.-H.G.), University of Helsinki, Finland
- Helsinki University Hospital, Research Program for Clinical and Molecular Metabolism, Faculty of Medicine (E.H.D., N.S., C.F., P.-H.G.), University of Helsinki, Finland
| | - Carol Forsblom
- Folkhälsan Institute of Genetics, Folkhälsan Research Center, Helsinki, Finland (E.H.D., N.S., C.F., P.-H.G.)
- Abdominal Center, Nephrology (E.H.D., N.S., C.F., P.-H.G.), University of Helsinki, Finland
- Helsinki University Hospital, Research Program for Clinical and Molecular Metabolism, Faculty of Medicine (E.H.D., N.S., C.F., P.-H.G.), University of Helsinki, Finland
| | - Anubha Mahajan
- Wellcome Centre for Human Genetics, Nuffield Department of Medicine (N.R.v.Z., M.A., A.M., N.R.R., N.W.R., M.I.M., A.P.M.), University of Oxford, United Kingdom
- Oxford Centre for Diabetes, Endocrinology and Metabolism, Radcliffe Department of Medicine (N.R.v.Z., A.M., N.R.R., N.W.R., M.I.M.), University of Oxford, United Kingdom
- Now with Genentech, South San Francisco, CA (A.M., M.I.M.)
| | - Neil R. Robertson
- Wellcome Centre for Human Genetics, Nuffield Department of Medicine (N.R.v.Z., M.A., A.M., N.R.R., N.W.R., M.I.M., A.P.M.), University of Oxford, United Kingdom
- Oxford Centre for Diabetes, Endocrinology and Metabolism, Radcliffe Department of Medicine (N.R.v.Z., A.M., N.R.R., N.W.R., M.I.M.), University of Oxford, United Kingdom
| | - N. William Rayner
- Wellcome Centre for Human Genetics, Nuffield Department of Medicine (N.R.v.Z., M.A., A.M., N.R.R., N.W.R., M.I.M., A.P.M.), University of Oxford, United Kingdom
- Oxford Centre for Diabetes, Endocrinology and Metabolism, Radcliffe Department of Medicine (N.R.v.Z., A.M., N.R.R., N.W.R., M.I.M.), University of Oxford, United Kingdom
- Department of Human Genetics, Wellcome Trust Sanger Institute, Hinxton, Cambridgeshire, United Kingdom (N.W.R.)
| | - Eero Lindholm
- Genomics, Diabetes and Endocrinology, Lund University Diabetes Centre, Malmö, Sweden (E. Ahlqvist, E.L., L.G.)
| | - Juha Sinisalo
- Heart and Lung Center (J.S.), University of Helsinki, Finland
| | - Markus Perola
- Institute for Molecular Medicine Finland (FIMM) (M.P., L.G.), University of Helsinki, Finland
- Finnish Institute for Health and Welfare, Helsinki, Finland (M.P., V.S.)
| | - Milla Kallio
- Vascular Surgery, Abdominal Center (M.K.), University of Helsinki, Finland
| | - Emily Weiss
- Usher Institute of Population Health Sciences and Informatics, The University of Edinburgh, United Kingdom (S.M., E.W., J.P.)
| | - Jackie Price
- Usher Institute of Population Health Sciences and Informatics, The University of Edinburgh, United Kingdom (S.M., E.W., J.P.)
| | - Andrew Paterson
- Dalla Lana School of Public Health, University of Toronto, ON, Canada (C.D.L., A.P.)
- Genetics & Genome Biology, SickKids, Toronto, ON, Canada (C.D.L., A.P.)
| | - Barbara Klein
- Ocular Epidemiology Research Group, University of Wisconsin-Madison (B.K.)
| | - Veikko Salomaa
- Finnish Institute for Health and Welfare, Helsinki, Finland (M.P., V.S.)
| | - Colin N.A. Palmer
- Pat Macpherson Centre for Pharmacogenetics and Pharmacogenomics, Ninewells Hospital and Medical School, University of Dundee, United Kingdom (C.N.A.P.)
| | - Per-Henrik Groop
- Folkhälsan Institute of Genetics, Folkhälsan Research Center, Helsinki, Finland (E.H.D., N.S., C.F., P.-H.G.)
- Abdominal Center, Nephrology (E.H.D., N.S., C.F., P.-H.G.), University of Helsinki, Finland
- Helsinki University Hospital, Research Program for Clinical and Molecular Metabolism, Faculty of Medicine (E.H.D., N.S., C.F., P.-H.G.), University of Helsinki, Finland
- Department of Medicine, Central Clinical School, Monash University, Melbourne, Victoria, Australia (P.-H.G.)
| | - Leif Groop
- Institute for Molecular Medicine Finland (FIMM) (M.P., L.G.), University of Helsinki, Finland
- Genomics, Diabetes and Endocrinology, Lund University Diabetes Centre, Malmö, Sweden (E. Ahlqvist, E.L., L.G.)
| | - Mark I. McCarthy
- Wellcome Centre for Human Genetics, Nuffield Department of Medicine (N.R.v.Z., M.A., A.M., N.R.R., N.W.R., M.I.M., A.P.M.), University of Oxford, United Kingdom
- Oxford Centre for Diabetes, Endocrinology and Metabolism, Radcliffe Department of Medicine (N.R.v.Z., A.M., N.R.R., N.W.R., M.I.M.), University of Oxford, United Kingdom
- Oxford NIHR Biomedical Research Centre, Oxford University Hospitals Trust, United Kingdom (M.I.M.)
- Now with Genentech, South San Francisco, CA (A.M., M.I.M.)
| | - Mariza de Andrade
- Department of Cardiovascular Medicine and the Gonda Vascular Center, Mayo Clinic, Rochester, MN (E. Austin, M.d.A., I.J.K.)
| | - Andrew P. Morris
- Wellcome Centre for Human Genetics, Nuffield Department of Medicine (N.R.v.Z., M.A., A.M., N.R.R., N.W.R., M.I.M., A.P.M.), University of Oxford, United Kingdom
- Department of Biostatistics, University of Liverpool, United Kingdom (A.P.M.)
- Centre for Genetics and Genomics Versus Arthritis, Centre for Musculoskeletal Research, The University of Manchester, United Kingdom (A.P.M.)
| | - Jemma C. Hopewell
- Clinical Trial Service Unit and Epidemiological Studies Unit, Nuffield Department of Population Health (A.S., J.C.H.), University of Oxford, United Kingdom
| | - Helen M. Colhoun
- Institute of Genetics and Molecular Medicine, University of Edinburgh, Western General Hospital Campus, United Kingdom (H.M.C.)
| | - Iftikhar J. Kullo
- Department of Cardiovascular Medicine and the Gonda Vascular Center, Mayo Clinic, Rochester, MN (E. Austin, M.d.A., I.J.K.)
| |
Collapse
|
13
|
von Eckardstein A. High Density Lipoproteins: Is There a Comeback as a Therapeutic Target? Handb Exp Pharmacol 2021; 270:157-200. [PMID: 34463854 DOI: 10.1007/164_2021_536] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Low plasma levels of High Density Lipoprotein (HDL) cholesterol (HDL-C) are associated with increased risks of atherosclerotic cardiovascular disease (ASCVD). In cell culture and animal models, HDL particles exert multiple potentially anti-atherogenic effects. However, drugs increasing HDL-C have failed to prevent cardiovascular endpoints. Mendelian Randomization studies neither found any genetic causality for the associations of HDL-C levels with differences in cardiovascular risk. Therefore, the causal role and, hence, utility as a therapeutic target of HDL has been questioned. However, the biomarker "HDL-C" as well as the interpretation of previous data has several important limitations: First, the inverse relationship of HDL-C with risk of ASCVD is neither linear nor continuous. Hence, neither the-higher-the-better strategies of previous drug developments nor previous linear cause-effect relationships assuming Mendelian randomization approaches appear appropriate. Second, most of the drugs previously tested do not target HDL metabolism specifically so that the futile trials question the clinical utility of the investigated drugs rather than the causal role of HDL in ASCVD. Third, the cholesterol of HDL measured as HDL-C neither exerts nor reports any HDL function. Comprehensive knowledge of structure-function-disease relationships of HDL particles and associated molecules will be a pre-requisite, to test them for their physiological and pathogenic relevance and exploit them for the diagnostic and therapeutic management of individuals at HDL-associated risk of ASCVD but also other diseases, for example diabetes, chronic kidney disease, infections, autoimmune and neurodegenerative diseases.
Collapse
Affiliation(s)
- Arnold von Eckardstein
- Institute of Clinical Chemistry, University Hospital Zurich and University of Zurich, Zurich, Switzerland.
| |
Collapse
|
14
|
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
|
15
|
Natorska J, Kopytek M, Undas A. Aortic valvular stenosis: Novel therapeutic strategies. Eur J Clin Invest 2021; 51:e13527. [PMID: 33621361 DOI: 10.1111/eci.13527] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Revised: 02/19/2021] [Accepted: 02/22/2021] [Indexed: 12/29/2022]
Abstract
BACKGROUND Aortic stenosis (AS) prevalence is estimated to reach 4.5 million cases worldwide by the year 2030. AS is a progressive disease without a pharmacological treatment. In the current review, we aimed to investigate novel therapeutic approaches for non-surgical AS treatment, at least in patients with mild-to-moderate AS. MATERIALS AND METHODS The most recent and relevant papers concerned with novel molecular pathways that have potential as therapeutic targets in AS were selected from searches of PubMed and Web of Science up to February 2021. RESULTS Growing evidence indicates that therapies using proprotein convertase subtilisin/kexin type 9 (PCSK9) inhibitors, simvastatin/ezetimibe combination, cholesteryl ester transfer protein inhibitors or antisense oligonucleotides targeting apolipoprotein(a) reduce the risk of AS progression. It has been shown that enhanced valvular lipid oxidation may drive AS development by leading to the activation of valvular interstitial cells (VICs), the most abundant valvular cells having a major contribution to valve calcification. Since VICs are able to release pro-inflammatory cytokines, clotting factors and proteins involved in calcification, strategies targeting these cell activations seem promising as therapeutic interventions. Recently, non-vitamin K antagonist oral anticoagulants (NOACs) have been shown to inhibit activation of VICs. CONCLUSION Several novel molecular pathways of AS development have been identified over the past few years. Therapies using PCSK9 inhibitors, simvastatin/ezetimibe combination, lipoprotein(a)-lowering therapy are highly promising candidates as therapeutics in the prevention of mild AS progression, while preclinical studies show that NOACs may inhibit valvular inflammation and coagulation activation and slower the rate of AS progression.
Collapse
Affiliation(s)
- Joanna Natorska
- John Paul II Hospital, Kraków, Poland.,Institute of Cardiology, Jagiellonian University Medical College, Kraków, Poland
| | - Magdalena Kopytek
- John Paul II Hospital, Kraków, Poland.,Institute of Cardiology, Jagiellonian University Medical College, Kraków, Poland
| | - Anetta Undas
- John Paul II Hospital, Kraków, Poland.,Institute of Cardiology, Jagiellonian University Medical College, Kraków, Poland
| |
Collapse
|
16
|
Beyond Lipoprotein(a) plasma measurements: Lipoprotein(a) and inflammation. Pharmacol Res 2021; 169:105689. [PMID: 34033878 PMCID: PMC9247870 DOI: 10.1016/j.phrs.2021.105689] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Revised: 05/18/2021] [Accepted: 05/19/2021] [Indexed: 12/20/2022]
Abstract
Genome wide association, epidemiological, and clinical studies have established high lipoprotein(a) [Lp(a)] as a causal risk factor for atherosclerotic cardiovascular disease (ASCVD). Lp(a) is an apoB100 containing lipoprotein covalently bound to apolipoprotein(a) [apo(a)], a glycoprotein. Plasma Lp(a) levels are to a large extent determined by genetics. Its link to cardiovascular disease (CVD) may be driven by its pro-inflammatory effects, of which its association with oxidized phospholipids (oxPL) bound to Lp(a) is the most studied. Various inflammatory conditions, such as rheumatoid arthritis (RA), systemic lupus erythematosus, acquired immunodeficiency syndrome, and chronic renal failure are associated with high Lp(a) levels. In cases of RA, high Lp(a) levels are reversed by interleukin-6 receptor (IL-6R) blockade by tocilizumab, suggesting a potential role for IL-6 in regulating Lp(a) plasma levels. Elevated levels of IL-6 and IL-6R polymorphisms are associated with CVD. Therapies aimed at lowering apo(a) and thereby reducing plasma Lp(a) levels are in clinical trials. Their results will determine if reductions in apo(a) and Lp(a) decrease cardiovascular outcomes. As we enter this new arena of available treatments, there is a need to improve our understanding of mechanisms. This review will focus on the role of Lp(a) in inflammation and CVD.
Collapse
|
17
|
Abstract
Abstract
Collapse
Affiliation(s)
- Angela Pirillo
- Center for the Study of Atherosclerosis, E. Bassini Hospital, Cinisello Balsamo, Milan, Italy
- IRCCS MultiMedica, Sesto S. Giovanni, Milan, Italy
| | - Alberico Luigi Catapano
- IRCCS MultiMedica, Sesto S. Giovanni, Milan, Italy
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Milan, Italy
| |
Collapse
|
18
|
Long term follow-up of genetically confirmed patients with familial hypercholesterolemia treated with first and second-generation statins and then with PCSK9 monoclonal antibodies. Atherosclerosis 2020; 308:6-14. [PMID: 32771803 DOI: 10.1016/j.atherosclerosis.2020.07.006] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Revised: 07/02/2020] [Accepted: 07/10/2020] [Indexed: 01/12/2023]
Abstract
BACKGROUND AND AIMS In Italy, the clinical and genetic characteristics of familial hypercholesterolemia (FH) have been extensively assessed in various lipid clinics, although no studies on long-term cardiovascular outcomes in heterozygous patients (He-FH) have been conducted. This study evaluated the incidence of atherosclerotic cardiovascular disease (ASCVD) in He-FH before and after a long-term period of lipid-lowering treatments to ascertain the interference of other risk factors. METHODS A total of 294 genetically characterised He-FH subjects from 1989 to 2019 were retrospectively analysed. General characteristics, lipid profiles, ASCVD prevalence, and ultrasound carotid atherosclerosis assessment were evaluated. Primary end points were ASCVD outcomes and the percentage of patients reaching recommended LDL-C targets. RESULTS During follow-up, despite a significant improvement in plasma lipid profiles, the ESC/EAS 2016 and 2019 recommended LDL cholesterol (LDL-C) goals were attained in only a few patients treated with anti-PCSK9 monoclonal antibodies added to the maximum tolerated oral therapy with statins plus ezetimibe. Forty-seven subjects had an ASCVD event before starting lipid-lowering therapy (LLT). During follow-up (median 13 years) on LLT, 28 patients had a first ASCVD event and 16 had recurrent ASCVD. In basal conditions and during follow-up, higher LDL-C levels were associated with increased ASCVD risk (p < 0.001). Prevention of recurrent ASCVD events was recorded with a long-term reduction of LDL-C below 100 mg/dl with statins plus ezetimibe. CONCLUSIONS PCSK9 inhibition is the only therapeutic option to achieve LDL-C goals as recommended for He-FH and can prevent ASCVD events as reported in large clinical trials. Long-term treatment with statins and ezetimibe seems to be effective at preventing ASCVD recurrence when LDL-C is maintained below 130 and 100 mg/dL for primary and secondary prevention, respectively.
Collapse
|
19
|
Greco MF, Sirtori CR, Corsini A, Ezhov M, Sampietro T, Ruscica M. Lipoprotein(a) Lowering-From Lipoprotein Apheresis to Antisense Oligonucleotide Approach. J Clin Med 2020; 9:jcm9072103. [PMID: 32635396 PMCID: PMC7408876 DOI: 10.3390/jcm9072103] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2020] [Revised: 06/24/2020] [Accepted: 07/01/2020] [Indexed: 12/11/2022] Open
Abstract
It is well-known that elevated lipoprotein(a)—Lp(a)—levels are associated with a higher risk of cardiovascular (CV) mortality and all-cause mortality, although a standard pharmacotherapeutic approach is still undefined for patients with high CV risk dependent on hyperlipoproteinemia(a). Combined with high Lp(a) levels, familial hypercholesterolemia (FH) leads to a greater CVD risk. In suspected FH patients, the proportion of cases explained by a rise of Lp(a) levels ranges between 5% and 20%. In the absence of a specific pharmacological approach able to lower Lp(a) to the extent required to achieve CV benefits, the most effective strategy today is lipoprotein apheresis (LA). Although limited, a clear effect on Lp(a) is exerted by PCSK9 antagonists, with apparently different mechanisms when given with statins (raised catabolism) or as monotherapy (reduced production). In the era of RNA-based therapies, a new dawn is represented by the use of antisense oligonucleotides APO(a)Lrx, able to reduce Lp(a) from 35% to over 80%, with generally modest injection site reactions. The improved knowledge of Lp(a) atherogenicity and possible prevention will be of benefit for patients with residual CV risk remaining after the most effective available lipid-lowering agents.
Collapse
Affiliation(s)
- Maria Francesca Greco
- Dipartimento di Science Farmacologiche e Biomolecolari, Università degli Studi di Milano, 20133 Milan, Italy; (M.F.G.); (A.C.)
| | - Cesare R. Sirtori
- Dyslipidemia Center, A.S.S.T. Grande Ospedale Metropolitano Niguarda, 20162 Milan, Italy;
| | - Alberto Corsini
- Dipartimento di Science Farmacologiche e Biomolecolari, Università degli Studi di Milano, 20133 Milan, Italy; (M.F.G.); (A.C.)
- IRCCS Multimedica, 20099 Milan, Italy
| | - Marat Ezhov
- National Medical Research Center of Cardiology of the Ministry of Health, Moscow, Russia;
| | - Tiziana Sampietro
- U.O. Lipoapheresis and Center for Inherited Dyslipidemias, Fondazione Toscana Gabriele Monasterio, 56126 Pisa, Italy;
| | - Massimiliano Ruscica
- Dipartimento di Science Farmacologiche e Biomolecolari, Università degli Studi di Milano, 20133 Milan, Italy; (M.F.G.); (A.C.)
- Correspondence: ; Tel.: +39-0250318220
| |
Collapse
|
20
|
Apolipoprotein(a), an enigmatic anti-angiogenic glycoprotein in human plasma: A curse or cure? Pharmacol Res 2020; 158:104858. [PMID: 32430285 DOI: 10.1016/j.phrs.2020.104858] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/18/2019] [Revised: 04/09/2020] [Accepted: 04/21/2020] [Indexed: 02/06/2023]
Abstract
Angiogenesis is a finely co-ordinated, multi-step developmental process of the new vascular structure. Even though angiogenesis is regularly occurring in physiological events such as embryogenesis, in adults, it is restricted to specific tissue sites where rapid cell-turnover and membrane synthesis occurs. Both excessive and insufficient angiogenesis lead to vascular disorders such as cancer, ocular diseases, diabetic retinopathy, atherosclerosis, intra-uterine growth restriction, ischemic heart disease, stroke etc. Occurrence of altered lipid profile and vascular lipid deposition along with vascular disorders is a hallmark of impaired angiogenesis. Among lipoproteins, lipoprotein(a) needs special attention due to the presence of a multi-kringle protein subunit, apolipoprotein(a) [apo(a)], which is structurally homologous to many naturally occurring anti-angiogenic proteins such as plasminogen and angiostatin. Researchers have constructed different recombinant forms of apo(a) (rhLK68, rhLK8, RHACK2, KV-11, and AU-6) and successfully exploited its potential to inhibit unwanted angiogenesis during tumor metastasis and retinal neovascularization. Similar to naturally occurring anti-angiogenic proteins, apo(a) can directly interfere with angiogenic signaling pathways. Besides this, apo(a) can also exert its anti-angiogenic effect indirectly by inducing endothelial cell apoptosis, by inhibiting endothelial progenitor cell functions or by upregulating nuclear factors in endothelial cells via apo(a)-bound oxPLs. However, the impact of the anti-angiogenic potential of native apo(a) during physiological angiogenesis in embryos and wounded tissues is not yet explored. In this context, we review the studies so far done to demonstrate the anti-angiogenic activity of apo(a) and the recent developments in using apo(a) as a therapeutic agent to treat impaired angiogenesis during vascular disorders, with emphasis on the gaps in the literature.
Collapse
|