551
|
Merzel Šabović EK, Starbek Zorko M, Janić M. Killing Two Birds with One Stone: Potential Therapies Targeting Psoriasis and Atherosclerosis at the Same Time. Int J Mol Sci 2022; 23:ijms23126648. [PMID: 35743091 PMCID: PMC9224172 DOI: 10.3390/ijms23126648] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Revised: 06/10/2022] [Accepted: 06/12/2022] [Indexed: 01/27/2023] Open
Abstract
Psoriasis is a chronic systemic inflammatory disease. Due to systemic inflammation, it is associated with many comorbidities. Among them, cardiovascular diseases represent the most common causes of morbidity and mortality in this population. Therefore, physicians treating patients with psoriasis should keep in mind that, as important as the treatment of psoriasis, awareness of cardiovascular risk deserves additional attention. Thus, in parallel with psoriasis treatment, a cardiovascular risk assessment must also be performed and addressed accordingly. In addition to encouraging non-pharmacologic strategies for a healthy lifestyle, physicians should be familiar with different pharmacologic options that can target psoriasis and reduce cardiovascular risk. In the present article, we present the pathophysiological mechanisms of the psoriasis and cardiometabolic interplay, our view on the interaction of psoriasis and cardiovascular disease, review the atherosclerotic effect of therapeutic options used in psoriasis, and vice versa, i.e., what the effect of medications used in the prevention of atherosclerosis could be on psoriasis.
Collapse
Affiliation(s)
- Eva Klara Merzel Šabović
- Department of Dermatovenerology, University Medical Centre Ljubljana, Gradiškova Ulica 10, SI-1000 Ljubljana, Slovenia;
- Faculty of Medicine, University of Ljubljana, Vrazov Trg 2, SI-1000 Ljubljana, Slovenia;
- Correspondence:
| | - Mateja Starbek Zorko
- Department of Dermatovenerology, University Medical Centre Ljubljana, Gradiškova Ulica 10, SI-1000 Ljubljana, Slovenia;
- Faculty of Medicine, University of Ljubljana, Vrazov Trg 2, SI-1000 Ljubljana, Slovenia;
| | - Miodrag Janić
- Faculty of Medicine, University of Ljubljana, Vrazov Trg 2, SI-1000 Ljubljana, Slovenia;
- Department of Endocrinology, Diabetes and Metabolic Diseases, University Medical Centre Ljubljana, Zaloška Cesta 7, SI-1000 Ljubljana, Slovenia
| |
Collapse
|
552
|
Anyfanti P, Margouta A, Goulas K, Gavriilaki M, Lazaridou E, Patsatsi A, Gkaliagkousi E. Endothelial Dysfunction in Psoriasis: An Updated Review. Front Med (Lausanne) 2022; 9:864185. [PMID: 35755028 PMCID: PMC9226899 DOI: 10.3389/fmed.2022.864185] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Accepted: 05/24/2022] [Indexed: 12/24/2022] Open
Abstract
Although psoriasis is predominantly a chronic inflammatory skin disorder, epidemiological data provide a solid link between psoriasis, especially in its more severe forms, and increased risk for cardiovascular morbidity and mortality. Apart from the increased prevalence of traditional cardiovascular risk factors, chronic inflammation appears to act synergistically with the underlying process of endothelial dysfunction toward the development of accelerated atherosclerosis, subclinical vascular injury and subsequently, clinically evident cardiovascular manifestations. Endothelial dysfunction is regarded as an early precursor of atherosclerosis with a predictive value for the development of future cardiovascular events. A thorough understanding of the mechanisms of endothelial dysfunction in psoriasis might pave the path for the development of more accurate cardiovascular risk prediction tools and possible therapeutic targets aiming to alleviate the increased cardiovascular burden associated with the disease. The present review summarizes the available evidence about the role of chronic inflammation and other important pathophysiological mechanisms involved in the development of endothelial dysfunction in psoriasis. An overview of studies implementing the most widely applied circulating and vascular biomarkers of endothelial dysfunction in psoriasis patients will be provided, and the impact of systemic psoriasis treatments on endothelial dysfunction and patients' cardiovascular risk will be discussed.
Collapse
Affiliation(s)
- Panagiota Anyfanti
- Department of Internal Medicine, Papageorgiou Hospital, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Anastasia Margouta
- Department of Internal Medicine, Papageorgiou Hospital, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Kyriakos Goulas
- Department of Internal Medicine, Papageorgiou Hospital, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Maria Gavriilaki
- Postgraduate Course, School of Medicine, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Elizabeth Lazaridou
- Department of Dermatology and Venereology, School of Medicine, Papageorgiou Hospital, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Aikaterini Patsatsi
- Department of Dermatology and Venereology, School of Medicine, Papageorgiou Hospital, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Eugenia Gkaliagkousi
- Department of Internal Medicine, Papageorgiou Hospital, Aristotle University of Thessaloniki, Thessaloniki, Greece
| |
Collapse
|
553
|
Gisterå A, Ketelhuth DFJ, Malin SG, Hansson GK. Animal Models of Atherosclerosis-Supportive Notes and Tricks of the Trade. Circ Res 2022; 130:1869-1887. [PMID: 35679358 DOI: 10.1161/circresaha.122.320263] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Atherosclerotic cardiovascular disease is a major cause of death among humans. Animal models have shown that cholesterol and inflammation are causatively involved in the disease process. Apolipoprotein B-containing lipoproteins elicit immune reactions and instigate inflammation in the vessel wall. Still, a treatment that is specific to vascular inflammation is lacking, which motivates continued in vivo investigations of the immune-vascular interactions that drive the disease. In this review, we distill old notions with emerging concepts into a contemporary understanding of vascular disease models. Pros and cons of different models are listed and the complex integrative interplay between cholesterol homeostasis, immune activation, and adaptations of the vascular system is discussed. Key limitations with atherosclerosis models are highlighted, and we suggest improvements that could accelerate progress in the field. However, excessively rigid experimental guidelines or limiting usage to certain animal models can be counterproductive. Continued work in improved models, as well as the development of new models, should be of great value in research and could aid the development of cardiovascular disease diagnostics and therapeutics of the future.
Collapse
Affiliation(s)
- Anton Gisterå
- Cardiovascular Medicine, Department of Medicine Solna, Karolinska Institutet and Center for Molecular Medicine, Karolinska University Hospital, Stockholm, Sweden (A.G., D.F.J.K., S.G.M., G.K.H.)
| | - Daniel F J Ketelhuth
- Cardiovascular Medicine, Department of Medicine Solna, Karolinska Institutet and Center for Molecular Medicine, Karolinska University Hospital, Stockholm, Sweden (A.G., D.F.J.K., S.G.M., G.K.H.).,Department of Cardiovascular and Renal Research, Institute for Molecular Medicine, University of Southern Denmark (SDU), Odense, Denmark (D.F.J.K)
| | - Stephen G Malin
- Cardiovascular Medicine, Department of Medicine Solna, Karolinska Institutet and Center for Molecular Medicine, Karolinska University Hospital, Stockholm, Sweden (A.G., D.F.J.K., S.G.M., G.K.H.)
| | - Göran K Hansson
- Cardiovascular Medicine, Department of Medicine Solna, Karolinska Institutet and Center for Molecular Medicine, Karolinska University Hospital, Stockholm, Sweden (A.G., D.F.J.K., S.G.M., G.K.H.)
| |
Collapse
|
554
|
Healthcare worker-based opportunistic screening for familial hypercholesterolemia in a low-resource setting. PLoS One 2022; 17:e0269605. [PMID: 35679249 PMCID: PMC9182245 DOI: 10.1371/journal.pone.0269605] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Accepted: 05/24/2022] [Indexed: 11/19/2022] Open
Abstract
Background & objective
Heterozygous familial hypercholesterolemia (FHeH) is important risk factor for premature coronary artery disease (CAD). Strategies for its diagnosis and prevalence have not been well studied in India. We performed healthcare worker-based opportunistic screening to assess feasibility for determining its prevalence.
Methods
A healthcare worker was trained in use of Dutch Lipid Clinic Network (DLCN) criteria for diagnosis of FHeH. Successive eligible individuals (n = 3000 of 3450 screened) presenting to biochemistry laboratories of two hospitals for blood lipid measurements were evaluated for FHeH. Cascade screening or genetic studies were not performed. Descriptive statistics are reported.
Results
We included 2549 participants (men 1870, women 679) not on statin therapy. Health worker screened 25–30 individuals/day in 6–10 minutes each. The mean age was 46.2±11y. Variables of DLCN criteria were more in women vs men: family history 51.1 vs 35.6%, past CAD 48.2 vs 20.1%, arcus cornealis 1.1 vs 0.3%, tendon xanthoma 0.3 vs 0.1%, and LDL cholesterol 190–249 mg/dl in 8.5 vs 2.4%, 250–329 mg/dl in 0.7 vs 0% and ≥330 mg/dl in 0.3 vs 0% (p<0.01). Definite FHeH (DLCN score >8) was in 15 (0.59%, frequency 1:170) and probable FHeH (score 6–8) in 87 (3.4%, frequency 1:29). The prevalence was significantly greater in women, age <50y and in those with hypertension, diabetes and known CAD.
Conclusions
Healthcare worker-led opportunistic screening for diagnosis of FHeH using DLCN criteria is feasible in low-resource settings. The results show significant prevalence of clinically detected definite and probable FHeH in the population studied.
Collapse
|
555
|
Mohd Nor NA, Budin SB, Zainalabidin S, Jalil J, Sapian S, Jubaidi FF, Mohamad Anuar NN. The Role of Polyphenol in Modulating Associated Genes in Diabetes-Induced Vascular Disorders. Int J Mol Sci 2022; 23:6396. [PMID: 35742837 PMCID: PMC9223817 DOI: 10.3390/ijms23126396] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2022] [Revised: 06/02/2022] [Accepted: 06/04/2022] [Indexed: 01/05/2023] Open
Abstract
Diabetes-induced vascular disorder is considered one of the deadly risk factors among diabetic patients that are caused by persistent hyperglycemia that eventually leads to cardiovascular diseases. Elevated reactive oxygen species (ROS) due to high blood glucose levels activate signaling pathways such as AGE/RAGE, PKC, polyol, and hexosamine pathways. The activated signaling pathway triggers oxidative stress, inflammation, and apoptosis which later lead to vascular dysfunction induced by diabetes. Polyphenol is a bioactive compound that can be found abundantly in plants such as vegetables, fruits, whole grains, and nuts. This compound exerts therapeutic effects in alleviating diabetes-induced vascular disorder, mainly due to its potential as an anti-oxidative, anti-inflammatory, and anti-apoptotic agent. In this review, we sought to summarize the recent discovery of polyphenol treatments in modulating associated genes involved in the progression of diabetes-induced vascular disorder.
Collapse
Affiliation(s)
- Nor Anizah Mohd Nor
- Centre for Diagnostic, Therapeutic and Investigative Studies, Faculty of Health Sciences, Universiti Kebangsaan Malaysia, Kuala Lumpur 50300, Malaysia; (N.A.M.N.); (S.B.B.); (S.S.); (F.F.J.)
- PICOMS International University College, Taman Batu Muda, Batu Caves, Kuala Lumpur 68100, Malaysia
| | - Siti Balkis Budin
- Centre for Diagnostic, Therapeutic and Investigative Studies, Faculty of Health Sciences, Universiti Kebangsaan Malaysia, Kuala Lumpur 50300, Malaysia; (N.A.M.N.); (S.B.B.); (S.S.); (F.F.J.)
| | - Satirah Zainalabidin
- Programme of Biomedical Science, Centre for Toxicology and Health Risk Research, Faculty of Health Sciences, Universiti Kebangsaan Malaysia, Kuala Lumpur 50300, Malaysia;
| | - Juriyati Jalil
- Center for Drug and Herbal Development, Faculty of Pharmacy, Universiti Kebangsaan Malaysia, Kuala Lumpur 50300, Malaysia;
| | - Syaifuzah Sapian
- Centre for Diagnostic, Therapeutic and Investigative Studies, Faculty of Health Sciences, Universiti Kebangsaan Malaysia, Kuala Lumpur 50300, Malaysia; (N.A.M.N.); (S.B.B.); (S.S.); (F.F.J.)
| | - Fatin Farhana Jubaidi
- Centre for Diagnostic, Therapeutic and Investigative Studies, Faculty of Health Sciences, Universiti Kebangsaan Malaysia, Kuala Lumpur 50300, Malaysia; (N.A.M.N.); (S.B.B.); (S.S.); (F.F.J.)
| | - Nur Najmi Mohamad Anuar
- Programme of Biomedical Science, Centre for Toxicology and Health Risk Research, Faculty of Health Sciences, Universiti Kebangsaan Malaysia, Kuala Lumpur 50300, Malaysia;
| |
Collapse
|
556
|
Grinshtein YI, Shabalin VV, Ruf RR, Shalnova SA, Drapkina OM. Atherogenic index of plasma as an additional marker of adverse cardiovascular outcomes. КАРДИОВАСКУЛЯРНАЯ ТЕРАПИЯ И ПРОФИЛАКТИКА 2022. [DOI: 10.15829/1728-8800-2022-3176] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022] Open
Abstract
Aim. Using a representative sample of the Krasnoyarsk Krai population, to evaluate the distribution of the atherogenic index of plasma (AIP), logarithmically transformed ratio of molar concentrations of triglycerides to high-density lipoprotein-cholesterol, and to identify possible associations between AIP, various cardiovascular diseases, and metabolic parameters.Material and methods. The study included a random representative sample of 1603 residents of the Krasnoyarsk and Berezovsky rural district aged 25-64 years as part of the Epidemiology of Cardiovascular Diseases and their Risk Factors in Regions of Russian Federation (ESSE-RF) study. Statistical processing was performed using IBM SPSS v22 and Microsoft Excel 2021 programs. We assessed the prevalence of hypertension (HTN), coronary artery disease, myocardial infarction (MI), stroke and renal dysfunction in the whole sample and in groups with different risk depending on AIP. Differences between groups were tested by Yates’s chi-squared test and were considered significant at p≤0,05.Results. Depending on AIP value, 73,5% of participants were in the low-risk group (AIP <0,10), 10,4% — in moderate risk group (AIP, 0,100,24) and 16,1% — in high-risk group (AIP>0,24). The prevalence of prior MI or stroke in the moderate and high-risk group for AIP was significantly higher than in the low-risk group (p=0,024). A regular increase in HTN prevalence was registered from 42,6% in the lowrisk group to 71,4% in the high-risk group for AIP (p<0,001 for all). There was a significant increase in the proportion of patients with a combination of HTN + elevated low-density lipoprotein cholesterol levels as AIP risk increased — from 28,4% at low risk to 45,2% at high risk. There were no significant differences between AIP risk groups in the prevalence of coronary artery disease and renal dysfunction.Conclusion. AIP is a simple additional estimated parameter that characterizes the atherogenic properties of plasma. Based on a cohort of Krasnoyarsk Krai subjects, an elevated level of AIP is associated with an increased prevalence of MI and strokes. Determination of AIP may be especially useful in the case of normal baseline low-density lipoprotein cholesterol levels.
Collapse
Affiliation(s)
| | - V. V. Shabalin
- V.F. Voyno-Yasenetsky Krasnoyarsk State Medical University
| | - R. R. Ruf
- V.F. Voyno-Yasenetsky Krasnoyarsk State Medical University
| | - S. A. Shalnova
- National Medical Research Center for Therapy and Preventive Medicine
| | - O. M. Drapkina
- National Medical Research Center for Therapy and Preventive Medicine
| |
Collapse
|
557
|
The LDL Apolipoprotein B-to-LDL Cholesterol Ratio: Association with Cardiovascular Mortality and a Biomarker of Small, Dense LDLs. Biomedicines 2022; 10:biomedicines10061302. [PMID: 35740324 PMCID: PMC9220033 DOI: 10.3390/biomedicines10061302] [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: 05/09/2022] [Accepted: 05/20/2022] [Indexed: 12/10/2022] Open
Abstract
Background and Objective: Small, dense low-density lipoproteins (LDLs) are considered more atherogenic than normal size LDLs. However, the measurement of small, dense LDLs requires sophisticated laboratory methods, such as ultracentrifugation, gradient gel electrophoresis, or nuclear magnetic resonance. We aimed to analyze whether the LDL apolipoprotein B (LDLapoB)-to-LDL cholesterol (LDLC) ratio is associated with cardiovascular mortality and whether this ratio represents a biomarker for small, dense LDLs. Methods: LDLC and LDLapoB were measured (beta-quantification) and calculated (according to Friedewald and Baca, respectively) for 3291 participants of the LURIC Study, with a median (inter-quartile range) follow-up for cardiovascular mortality of 9.9 (8.7−10.7) years. An independent replication cohort included 1660 participants. Associations of the LDLapoB/LDLC ratio with LDL subclass particle concentrations (ultracentrifugation) were tested for 282 participants. Results: In the LURIC Study, the mean (standard deviation) LDLC and LDLapoB concentrations were 117 (34) and 85 (22) mg/dL, respectively; 621 cardiovascular deaths occurred. Elevated LDLapoB/LDLC (calculated and measured) ratios were significantly and independently associated with increased cardiovascular mortality in the entire cohort (fourth vs. first quartile: hazard ratio (95% confidence interval) = 2.07 (1.53−2.79)) and in statin-naïve patients. The association between calculated LDLapoB/LDLC ratio and cardiovascular mortality was replicated in an independent cohort. High LDLapoB/LDLC ratios were associated with higher LDL5 and LDL6 concentrations (both p < 0.001), but not with concentrations of larger LDLs. Conclusions: Elevated measured and calculated LDLapoB/LDLC ratios are associated with increased cardiovascular mortality. Use of LDLapoB/LDLC ratios allows estimation of the atherogenic risk conferred by small, dense LDLs.
Collapse
|
558
|
Glise L, Rutberg M, Håversen L, Levin MC, Levin M, Jeppsson A, Borén J, Fogelstrand P. pH-Dependent Protonation of Histidine Residues Is Critical for Electrostatic LDL (Low-Density Lipoprotein) Binding to Human Coronary Arteries. Arterioscler Thromb Vasc Biol 2022; 42:1037-1047. [PMID: 35652335 DOI: 10.1161/atvbaha.122.317868] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND The initiating step in atherogenesis is the electrostatic binding of LDL (low-density lipoprotein) to proteoglycan glycosaminoglycans in the arterial intima. However, although proteoglycans are widespread throughout the intima of most coronary artery segments, LDL is not evenly distributed, indicating that LDL retention is not merely dependent on the presence of proteoglycans. We aim to identify factors that promote the interaction between LDL and the vessel wall of human coronary arteries. METHODS We developed an ex vivo model to investigate binding of human-labeled LDL to human coronary artery sections without the interference of cellular processes. RESULTS By staining consecutive sections of human coronary arteries, we found strong staining of sulfated glycosaminoglycans throughout the arterial intima, whereas endogenous LDL deposits were focally distributed. Ex vivo binding of LDL was uniform in all intimal areas with sulfated glycosaminoglycans. However, lowering the pH from 7.4 to 6.5 triggered a 35-fold increase in LDL binding. The pH-dependent binding was abolished by pretreating LDL with diethyl-pyrocarbonate, which blocks the protonation of histidine residues, or cyclohexanedione, which inhibits the positive charge of site B on LDL. Thus, both histidine protonation and site B are required for strong electrostatic LDL binding to the intima. CONCLUSIONS This study identifies histidine protonation as an important component for electrostatic LDL binding to human coronary arteries. Our findings show that the local pH will have a profound impact on LDL's affinity for sulfated glycosaminoglycans, which may influence the retention and accumulation pattern of LDL in the arterial vasculature.
Collapse
Affiliation(s)
- Lars Glise
- Department of Molecular and Clinical Medicine, Institute of Medicine, University of Gothenburg, Sweden (L.G., M.R., L.H., M.C.L., M.L., A.J., J.B., P.F.)
| | - Mikael Rutberg
- Department of Molecular and Clinical Medicine, Institute of Medicine, University of Gothenburg, Sweden (L.G., M.R., L.H., M.C.L., M.L., A.J., J.B., P.F.)
| | - Liliana Håversen
- Department of Molecular and Clinical Medicine, Institute of Medicine, University of Gothenburg, Sweden (L.G., M.R., L.H., M.C.L., M.L., A.J., J.B., P.F.)
| | - Malin C Levin
- Department of Molecular and Clinical Medicine, Institute of Medicine, University of Gothenburg, Sweden (L.G., M.R., L.H., M.C.L., M.L., A.J., J.B., P.F.)
| | - Max Levin
- Department of Molecular and Clinical Medicine, Institute of Medicine, University of Gothenburg, Sweden (L.G., M.R., L.H., M.C.L., M.L., A.J., J.B., P.F.)
| | - Anders Jeppsson
- Department of Molecular and Clinical Medicine, Institute of Medicine, University of Gothenburg, Sweden (L.G., M.R., L.H., M.C.L., M.L., A.J., J.B., P.F.).,Department of Cardiothoracic Surgery, Sahlgrenska University Hospital, Gothenburg, Sweden (A.J.)
| | - Jan Borén
- Department of Molecular and Clinical Medicine, Institute of Medicine, University of Gothenburg, Sweden (L.G., M.R., L.H., M.C.L., M.L., A.J., J.B., P.F.).,Sahlgrenska University Hospital/Wallenberg Laboratory, Gothenburg, Sweden (J.B.)
| | - Per Fogelstrand
- Department of Molecular and Clinical Medicine, Institute of Medicine, University of Gothenburg, Sweden (L.G., M.R., L.H., M.C.L., M.L., A.J., J.B., P.F.)
| |
Collapse
|
559
|
Abstract
PURPOSE OF REVIEW Levels of small, dense low-density lipoprotein (LDL) (sdLDL) particles determined by several analytic procedures have been associated with risk of atherosclerotic cardiovascular disease (ASCVD). This review focuses on the clinical significance of sdLDL measurement. RECENT FINDINGS Results of multiple prospective studies have supported earlier evidence that higher levels of sdLDL are significantly associated with greater ASCVD risk, in many cases independent of other lipid and ASCVD risk factors as well as levels of larger LDL particles. A number of properties of sdLDL vs. larger LDL, including reduced LDL receptor affinity and prolonged plasma residence time as well as greater oxidative susceptibility and affinity for arterial proteoglycans, are consistent with their heightened atherogenic potential. Nevertheless, determination of the extent to which sdLDL can preferentially impact ASCVD risk compared with other apoprotein B-containing lipoproteins has been confounded by their metabolic interrelationships and statistical collinearity, as well as differences in analytic procedures and definitions of sdLDL. SUMMARY A growing body of data points to sdLDL concentration as a significant determinant of ASCVD risk. Although future studies should be aimed at determining the clinical benefit of reducing sdLDL levels, there is sufficient evidence to warrant consideration of sdLDL measurement in assessing and managing risk of cardiovascular disease. VIDEO ABSTRACT https://www.dropbox.com/s/lioohr2ead7yx2p/zoom_0.mp4?dl=0.
Collapse
|
560
|
Phillips MC. Metabolic Strategies in Healthcare: A New Era. Aging Dis 2022; 13:655-672. [PMID: 35656107 PMCID: PMC9116908 DOI: 10.14336/ad.2021.1018] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2021] [Accepted: 10/18/2021] [Indexed: 12/14/2022] Open
Abstract
Modern healthcare systems are founded on a disease-centric paradigm, which has conferred many notable successes against infectious disorders in the past. However, today's leading causes of death are dominated by non-infectious "lifestyle" disorders, broadly represented by the metabolic syndrome, atherosclerosis, cancer, and neurodegeneration. Our disease-centric paradigm regards these disorders as distinct disease processes, caused and driven by disease targets that must be suppressed or eliminated to clear the disease. By contrast, a health-centric paradigm recognizes the lifestyle disorders as a series of hormonal and metabolic responses to a singular, lifestyle-induced disease of mitochondria dysfunction, a disease target that must be restored to improve health, which may be defined as optimized mitochondria function. Seen from a health-centric perspective, most drugs target a response rather than the disease, whereas metabolic strategies, such as fasting and carbohydrate-restricted diets, aim to restore mitochondria function, mitigating the impetus that underlies and drives the lifestyle disorders. Substantial human evidence indicates either strategy can effectively mitigate the metabolic syndrome. Preliminary evidence also indicates potential benefits in atherosclerosis, cancer, and neurodegeneration. Given the existing evidence, integrating metabolic strategies into modern healthcare systems should be identified as a global health priority.
Collapse
|
561
|
Teerakanchana’s equation transcends over 12 other LDL-C quantification formulae in the North Indian population. Clin Chim Acta 2022; 531:168-176. [DOI: 10.1016/j.cca.2022.04.006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Revised: 04/02/2022] [Accepted: 04/05/2022] [Indexed: 01/06/2023]
|
562
|
Libby P, Tokgözoğlu L. Chasing LDL cholesterol to the bottom - PCSK9 in perspective. NATURE CARDIOVASCULAR RESEARCH 2022; 1:554-561. [PMID: 39195874 DOI: 10.1038/s44161-022-00085-x] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Accepted: 05/10/2022] [Indexed: 08/29/2024]
Abstract
Low-density lipoprotein (LDL) indubitably contributes causally to atherosclerosis, a leading challenge to health worldwide. Interventions that lower LDL cholesterol (LDL-C) have made remarkable inroads against this global scourge. Recent therapeutic advances have achieved ever lower levels of LDL-C. Improved cardiovascular outcomes continue to accrue from these interventions. In particular, the discovery of the role of proprotein convertase subtilisin/kexin type 9 (PCSK9) as the causal gene in autosomal-dominant hypercholesterolemia has led with remarkable speed to the development of novel agents to lower LDL-C concentrations beyond prior measures, and to alleviate further cardiovascular risk. We review how this story, and its position in the broader landscape of therapy to prevent atherosclerotic events, represents a notable victory of contemporary cardiovascular medicine and reflects successful partnerships between basic scientists, the pharmaceutical and biotechnology sectors, and clinical investigators. Continued cooperation in this manner promises to yield further progress in combating cardiovascular diseases beyond interventions on LDL-C.
Collapse
Affiliation(s)
- Peter Libby
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
| | - Lale Tokgözoğlu
- Department of Cardiology, Hacettepe University Faculty of Medicine, Ankara, Turkey
| |
Collapse
|
563
|
Dar GM, Dash M, Mahajan B. Teerakanchana’s equation transcends over 12 other LDL-C quantification formulae in the North Indian population. Clin Chim Acta 2022; 531:168-176. [DOI: https:/doi.org/10.1016/j.cca.2022.04.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/28/2023]
|
564
|
Opportunità cliniche e impatto sul sistema sanitario di un trattamento ottimale del paziente post-sindrome coronarica acuta. GLOBAL & REGIONAL HEALTH TECHNOLOGY ASSESSMENT 2022; 9:17-26. [PMID: 36628067 PMCID: PMC9796606 DOI: 10.33393/grhta.2022.2391] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Accepted: 03/23/2022] [Indexed: 01/13/2023] Open
Abstract
Despite the improvement of revascularization procedures, patients with acute coronary syndrome often develop recurrent ischemic events, suggesting a high residual cardiovascular risk in these patients, which requires a strict clinical monitoring as well as an optimal control of modifiable risk factors. To this aim, an optimal management of index event and appropriate preventive measures are equally important. Hospital care by cardiologists should be followed by outpatient management by general practitioners, as established by specific diagnostic and therapeutic pathways, which should warrant an optimal support to the patient. A strict collaboration between hospital and primary care is crucial to monitor and adapt drug therapy after the acute event and improve adherence of the patients to prescribed treatments and implementation of life-style modifications, with benefits also in term of cost-effectiveness. In this context, individualized rehabilitation programs should also be offered to patients with acute coronary syndromes, in order to improve survival and quality of life.
Collapse
|
565
|
Cipryan L, Litschmannova M, Maffetone PB, Plews DJ, Dostal T, Hofmann P, Laursen PB. Very Low-Carbohydrate High-Fat Diet Improves Risk Markers for Cardiometabolic Health More Than Exercise in Men and Women With Overfat Constitution: Secondary Analysis of a Randomized Controlled Clinical Trial. Front Nutr 2022; 9:867690. [PMID: 35677551 PMCID: PMC9168912 DOI: 10.3389/fnut.2022.867690] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Accepted: 04/29/2022] [Indexed: 12/15/2022] Open
Abstract
Purpose This randomized controlled parallel-group study examined the effects of a very low-carbohydrate high-fat (VLCHF) diet and high-intensity interval training (HIIT) program over 12-weeks on cardiometabolic risk factors in individuals with overfat constitution. Methods Ninety-one participants out of 109 completed the study. The participants were randomly allocated to the HIIT (N = 22), VLCHF (N = 25), VLCHF+HIIT (N = 25), or control (N = 19) groups for 12 weeks. Fasting plasma samples were collected before the intervention and after 4 and 12 weeks. The analyzed outcomes included complete blood count, glucose, insulin, glycated hemoglobin, triglycerides (TG), cholesterol, high- and low-density lipoprotein (HDL-C and LDL-C), lipoprotein(a), adiponectin (Adpn), leptin (Lep), tumor necrosis factor α (TNF-α), other interleukins (hs-IL-6, IL-1β, and IL-10), and IL-1RA. The homeostasis model assessment of insulin resistance (HOMA-IR), Adpn/Lep ratio, TG/HDL-C ratio, and TyG index were calculated and analyzed. Blood pressure was measured before the intervention, after 4, 8, and 12 weeks (ClinicalTrials.gov: NCT03934476). Results Absolute changes in HOMA-IR, Adpn/Lep ratio, LDL-C, and diastolic blood pressure after 12 weeks differed by study groups (p < 0.05). The most pronounced changes were revealed in the VLCHF (ΔM [95% CI]; HOMA-IR: -0.75 [-1.13; -0.55]; Adpn/Lep: 9.34 [6.33; 37.39]; LDL-C: 0.06 [-0.12; 0.50] mmol/l) and VLCHF+HIIT (HOMA-IR: -0.44 [-1.14; 0.12]; Adpn/Lep: 4.26 [2.24; 13.16]; LDL-C: 0.25 [-0.04; 0.50] mmol/l) groups. Conclusions A 12-week VLCHF diet intervention in individuals with overfat constitution is effective for favorable changes in HOMA-IR (compared to HIIT), Adpn/Lep ratio, and diastolic blood pressure. HIIT, or HIIT combined with the VLCHF diet, had no additional benefits for the analyzed variables. No adverse side effects were observed.
Collapse
Affiliation(s)
- Lukas Cipryan
- Department of Human Movement Studies & Human Motion Diagnostic Centre, The University of Ostrava, Ostrava, Czechia
| | - Martina Litschmannova
- Department of Applied Mathematics, VSB—Technical University of Ostrava, Ostrava, Czechia
| | | | - Daniel J. Plews
- Sports Performance Research Institute New Zealand (SPRINZ), Auckland University of Technology, Auckland, New Zealand
| | - Tomas Dostal
- Department of Human Movement Studies & Human Motion Diagnostic Centre, The University of Ostrava, Ostrava, Czechia
| | - Peter Hofmann
- Institute of Human Movement Science, Sport & Health, Exercise Physiology, Training & Training Therapy Research Group, University of Graz, Graz, Austria
| | - Paul B. Laursen
- Sports Performance Research Institute New Zealand (SPRINZ), Auckland University of Technology, Auckland, New Zealand
| |
Collapse
|
566
|
Exploring Endothelial Colony-Forming Cells to Better Understand the Pathophysiology of Disease: An Updated Review. Stem Cells Int 2022; 2022:4460041. [PMID: 35615696 PMCID: PMC9126670 DOI: 10.1155/2022/4460041] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Revised: 04/20/2022] [Accepted: 05/09/2022] [Indexed: 12/12/2022] Open
Abstract
Endothelial cell (EC) dysfunction has been implicated in a variety of pathological conditions. The collection of ECs from patients is typically conducted postmortem or through invasive procedures, such as surgery and interventional procedures, hampering efforts to clarify the role of ECs in disease onset and progression. In contrast, endothelial colony-forming cells (ECFCs), also termed late endothelial progenitor cells, late outgrowth endothelial cells, blood outgrowth endothelial cells, or endothelial outgrowth cells, are obtained in a minimally invasive manner, namely, by the culture of human peripheral blood mononuclear cells in endothelial growth medium. ECFCs resemble mature ECs phenotypically, genetically, and functionally, making them excellent surrogates for ECs. Numerous studies have been performed that examined ECFC function in conditions such as coronary artery disease, diabetes mellitus, hereditary hemorrhagic telangiectasia, congenital bicuspid aortic valve disease, pulmonary arterial hypertension, venous thromboembolic disease, and von Willebrand disease. Here, we provide an updated review of studies using ECFCs that were performed to better understand the pathophysiology of disease. We also discuss the potential of ECFCs as disease biomarkers and the standardized methods to culture, quantify, and evaluate ECFCs and suggest the future direction of research in this field.
Collapse
|
567
|
Xiang Q, Tian F, Xu J, Du X, Zhang S, Liu L. New insight into dyslipidemia‐induced cellular senescence in atherosclerosis. Biol Rev Camb Philos Soc 2022; 97:1844-1867. [PMID: 35569818 PMCID: PMC9541442 DOI: 10.1111/brv.12866] [Citation(s) in RCA: 59] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Revised: 04/18/2022] [Accepted: 04/28/2022] [Indexed: 11/28/2022]
Abstract
Atherosclerosis, characterized by lipid‐rich plaques in the arterial wall, is an age‐related disorder and a leading cause of mortality worldwide. However, the specific mechanisms remain complex. Recently, emerging evidence has demonstrated that senescence of various types of cells, such as endothelial cells (ECs), vascular smooth muscle cells (VSMCs), macrophages, endothelial progenitor cells (EPCs), and adipose‐derived mesenchymal stem cells (AMSCs) contributes to atherosclerosis. Cellular senescence and atherosclerosis share various causative stimuli, in which dyslipidemia has attracted much attention. Dyslipidemia, mainly referred to elevated plasma levels of atherogenic lipids or lipoproteins, or functional impairment of anti‐atherogenic lipids or lipoproteins, plays a pivotal role both in cellular senescence and atherosclerosis. In this review, we summarize the current evidence for dyslipidemia‐induced cellular senescence during atherosclerosis, with a focus on low‐density lipoprotein (LDL) and its modifications, hydrolysate of triglyceride‐rich lipoproteins (TRLs), and high‐density lipoprotein (HDL), respectively. Furthermore, we describe the underlying mechanisms linking dyslipidemia‐induced cellular senescence and atherosclerosis. Finally, we discuss the senescence‐related therapeutic strategies for atherosclerosis, with special attention given to the anti‐atherosclerotic effects of promising geroprotectors as well as anti‐senescence effects of current lipid‐lowering drugs.
Collapse
Affiliation(s)
- Qunyan Xiang
- Department of Geriatrics, The Second Xiangya Hospital Central South University Changsha Hunan 410011 PR China
- Institute of Aging and Age‐related Disease Research Central South University Changsha Hunan 410011 PR China
| | - Feng Tian
- Department of Geriatric Cardiology The First Affiliated Hospital of Zhengzhou University Zhengzhou Henan 450000 PR China
| | - Jin Xu
- Department of Cardiovascular Medicine, The Second Xiangya Hospital Central South University Changsha Hunan 410011 PR China
- Research Institute of Blood Lipid and Atherosclerosis Central South University Changsha Hunan 410011 PR China
- Modern Cardiovascular Disease Clinical Technology Research Center of Hunan Province Changsha Hunan 410011 PR China
- Cardiovascular Disease Research Center of Hunan Province Changsha Hunan 410011 PR China
| | - Xiao Du
- Department of Cardiovascular Medicine, The Second Xiangya Hospital Central South University Changsha Hunan 410011 PR China
- Research Institute of Blood Lipid and Atherosclerosis Central South University Changsha Hunan 410011 PR China
- Modern Cardiovascular Disease Clinical Technology Research Center of Hunan Province Changsha Hunan 410011 PR China
- Cardiovascular Disease Research Center of Hunan Province Changsha Hunan 410011 PR China
| | - Shilan Zhang
- Department of Gastroenterology, The Second Xiangya Hospital Central South University Changsha Hunan 410011 PR China
| | - Ling Liu
- Department of Cardiovascular Medicine, The Second Xiangya Hospital Central South University Changsha Hunan 410011 PR China
- Research Institute of Blood Lipid and Atherosclerosis Central South University Changsha Hunan 410011 PR China
- Modern Cardiovascular Disease Clinical Technology Research Center of Hunan Province Changsha Hunan 410011 PR China
- Cardiovascular Disease Research Center of Hunan Province Changsha Hunan 410011 PR China
| |
Collapse
|
568
|
Georgakis MK, Bernhagen J, Heitman LH, Weber C, Dichgans M. Targeting the CCL2-CCR2 axis for atheroprotection. Eur Heart J 2022; 43:1799-1808. [PMID: 35567558 DOI: 10.1093/eurheartj/ehac094] [Citation(s) in RCA: 85] [Impact Index Per Article: 28.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2021] [Revised: 12/22/2021] [Accepted: 02/15/2022] [Indexed: 11/12/2022] Open
Abstract
Decades of research have established atherosclerosis as an inflammatory disease. Only recently though, clinical trials provided proof-of-concept evidence for the efficacy of anti-inflammatory strategies with respect to cardiovascular events, thus offering a new paradigm for lowering residual vascular risk. Efforts to target the inflammasome-interleukin-1β-interleukin-6 pathway have been highly successful, but inter-individual variations in drug response, a lack of reduction in all-cause mortality, and a higher rate of infections also highlight the need for a second generation of anti-inflammatory agents targeting atherosclerosis-specific immune mechanisms while minimizing systemic side effects. CC-motif chemokine ligand 2/monocyte-chemoattractant protein-1 (CCL2/MCP-1) orchestrates inflammatory monocyte trafficking between the bone marrow, circulation, and atherosclerotic plaques by binding to its cognate receptor CCR2. Adding to a strong body of data from experimental atherosclerosis models, a coherent series of recent large-scale genetic and observational epidemiological studies along with data from human atherosclerotic plaques highlight the relevance and therapeutic potential of the CCL2-CCR2 axis in human atherosclerosis. Here, we summarize experimental and human data pinpointing the CCL2-CCR2 pathway as an emerging drug target in cardiovascular disease. Furthermore, we contextualize previous efforts to interfere with this pathway, scrutinize approaches of ligand targeting vs. receptor targeting, and discuss possible pathway-intrinsic opportunities and challenges related to pharmacological targeting of the CCL2-CCR2 axis in human atherosclerotic disease.
Collapse
Affiliation(s)
- Marios K Georgakis
- Institute for Stroke and Dementia Research (ISD), University Hospital, LMU Munich, D-81377 Munich, Germany
- Center of Genomic Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Jürgen Bernhagen
- Institute for Stroke and Dementia Research (ISD), University Hospital, LMU Munich, D-81377 Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance, Munich, Germany
| | - Laura H Heitman
- Division of Drug Discovery and Safety, Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands
- Oncode Institute, Utrecht, The Netherlands
| | - Christian Weber
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance, Munich, Germany
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians-Universität (LMU) Munich, Germany
- Institute for Genetic and Biomedical Research, UoS of Milan, National Research Council, Milan, Italy
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, The Netherlands
| | - Martin Dichgans
- Institute for Stroke and Dementia Research (ISD), University Hospital, LMU Munich, D-81377 Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
- German Centre for Neurodegenerative Diseases (DZNE), Munich, Germany
| |
Collapse
|
569
|
Atherogenic Plasma Index or Non-High-Density Lipoproteins as Markers Best Reflecting Age-Related High Concentrations of Small Dense Low-Density Lipoproteins. Int J Mol Sci 2022; 23:ijms23095089. [PMID: 35563477 PMCID: PMC9102874 DOI: 10.3390/ijms23095089] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Revised: 04/19/2022] [Accepted: 04/29/2022] [Indexed: 02/07/2023] Open
Abstract
The study aimed to assess the strength of the relationships between small dense low-density lipoproteins (sdLDL) and other parameters describing metabolic disorders and determine which of the lipid profile parameters can be used as markers of increased sdLDL concentration. The proposed model of sdLDL (examined by heparin−magnesium precipitation method) as a function of lipid parameters and atherogenic plasma indexes non-high-dense lipoproteins (non-HDL) and total cholesterol to high-dense lipoprotein ratio (TC/HDL), Atherogenic plasma index (API) is based on data from 485 participants divided into two age groups, <35≥ years. In multiple linear regression, sdLDL concentration was associated with the concentration of non-HDL-C (p = 0.043) and API value (p < 0.001) in participants <35 years, and with non-HDL-C (p < 0.001) and triglycerides (p = 0.020) concentration ≥35 years. The presence of abnormal values of API in participants <35 years and non-HDL-C in participants ≥35 years is a significant factor increasing the chances of the highest sdLDL (≥1.03 mmol/L) corresponding to Q4 in people without metabolic disorders. Different lipid parameters and atherogenicity indexes are associated with a high concentration of sdLDL depending on the age group. Abnormal API <35 years and non-HDL ≥35 years are associated with the highest sdLDL values and may be an indication for further specialist diagnosis of cardiovascular disease risk factors.
Collapse
|
570
|
Sekimoto T, Koba S, Mori H, Arai T, Matsukawa N, Sakai R, Yokota Y, Sato S, Tanaka H, Masaki R, Oishi Y, Ogura K, Arai K, Nomura K, Sakai K, Tsujita H, Kondo S, Tsukamoto S, Matsumoto H, Suzuki H, Shinke T. Impact of Small Dense Low-Density Lipoprotein Cholesterol on Cholesterol Crystals in Patients with Acute Coronary Syndrome: An Optical Coherence Tomography Study. J Clin Lipidol 2022; 16:438-446. [DOI: 10.1016/j.jacl.2022.04.008] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Revised: 04/08/2022] [Accepted: 04/28/2022] [Indexed: 11/24/2022]
|
571
|
Toth PP, Chapman MJ, Parhofer KG, Nelson JR. Differentiating EPA from EPA/DHA in cardiovascular risk reduction. AMERICAN HEART JOURNAL PLUS : CARDIOLOGY RESEARCH AND PRACTICE 2022; 17:100148. [PMID: 38559888 PMCID: PMC10978325 DOI: 10.1016/j.ahjo.2022.100148] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Revised: 05/19/2022] [Accepted: 05/19/2022] [Indexed: 04/04/2024]
Abstract
None of the clinical trials of omega-3 fatty acids using combinations of eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA) were able to show any effect on cardiovascular outcomes, despite reductions in triglyceride levels. In contrast, the Reduction of Cardiovascular Events With Icosapent Ethyl-Intervention Trial (REDUCE-IT), which employed high-dose (4 g) purified EPA, demonstrated a 25% reduction in atherosclerotic cardiovascular disease-related events compared with placebo (hazard ratio 0.75; 95% confidence interval 0.68-0.83; P < 0.001). Moreover, REDUCE-IT is the first clinical trial using a lipid-lowering agent as adjuvant therapy to a statin to show a significant reduction in cardiovascular mortality. Significant reductions in stroke, need for revascularization, and myocardial infarction were also observed. The pharmacology of EPA is distinct from that of DHA, with a differential effect on membrane structure, lipoprotein oxidation, and the production of downstream metabolites that promote the resolution of inflammation. Attained plasma levels of EPA may be an important determinant of efficacy, with a substudy of REDUCE-IT suggesting that the threshold for clinical benefit of EPA is approximately 100 μg/mL, a level achieved in only a minority of patients in other studies. No similar clinical trials of DHA monotherapy have been conducted, so no such threshold has been established. The results of the REDUCE-IT and the Japan EPA Lipid Intervention Study (JELIS) together affirm the efficacy of EPA therapy for cardiovascular disease risk reduction in certain patient populations.
Collapse
Affiliation(s)
- Peter P. Toth
- CGH Medical Center, Sterling, IL, USA
- Cicarrone Center for the Prevention of Cardiovascular Disease, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | | | - Klaus G. Parhofer
- Medical Clinic IV – Grosshadern Hospital of the University of Munich, Munich, Germany
| | | |
Collapse
|
572
|
Mallat Z, Binder CJ. The why and how of adaptive immune responses in ischemic cardiovascular disease. NATURE CARDIOVASCULAR RESEARCH 2022; 1:431-444. [PMID: 36382200 PMCID: PMC7613798 DOI: 10.1038/s44161-022-00049-1] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Accepted: 03/10/2022] [Indexed: 02/02/2023]
Abstract
Atherosclerotic cardiovascular disease is a major cause of disability and death worldwide. Most therapeutic approaches target traditional risk factors but ignore the fundamental role of the immune system. This is a huge unmet need. Recent evidence indicates that reducing inflammation may limit cardiovascular events. However, the concomitant increase in the risk of lifethreatening infections is a major drawback. In this context, targeting adaptive immunity could constitute a highly effective and safer approach. In this Review, we address the why and how of the immuno-cardiovascular unit, in health and in atherosclerotic disease. We review and discuss fundamental mechanisms that ensure immune tolerance to cardiovascular tissue, and examine how their disruption promotes disease progression. We identify promising strategies to manipulate the adaptive immune system for patient benefit, including novel biologics and RNA-based vaccination strategies. Finally, we advocate for establishing a molecular classification of atherosclerosis as an important milestone in our quest to radically change the understanding and treatment of atherosclerotic disease.
Collapse
Affiliation(s)
- Ziad Mallat
- Division of Cardiovascular Medicine, Department of Medicine, University of Cambridge, Cambridge, UK
- Unversité de Paris, and INSERM U970, Paris Cardiovascular Research Centre, Paris, France
| | - Christoph J. Binder
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
573
|
Cimmino G, Di Serafino L, Cirillo P. Pathophysiology and mechanisms of Acute Coronary Syndromes: atherothrombosis, immune-inflammation, and beyond. Expert Rev Cardiovasc Ther 2022; 20:351-362. [PMID: 35510629 DOI: 10.1080/14779072.2022.2074836] [Citation(s) in RCA: 4] [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: 01/26/2022] [Accepted: 05/04/2022] [Indexed: 12/24/2022]
Abstract
INTRODUCTION The pathophysiology of atherosclerosis and its acute complications, such as the Acute Coronary Syndromes (ACS), is continuously under investigation. Immunity and inflammation seem to play a pivotal role in promoting formation and grow of atherosclerotic plaques. At the same time, plaque rupture followed by both platelets' activation and coagulation cascade induction lead to intracoronary thrombus formation. Although these phenomena might be considered responsible of about 90% of ACS, in up to 5-10% of acute syndromes, a non-obstructive coronary artery disease (MINOCA) might be documented. This paper gives an overview on atherothrombosis and immuno-inflammation processes involved in ACS pathophysiology, also emphasizing the pathological mechanisms potentially involved in MINOCA. AREAS COVERED The relationship between immuno-inflammation and atherothrombosis is continuously updated by recent findings. At the same time, pathophysiology of MINOCA still remains a partially unexplored field, stimulating the research of potential links between these two aspects of ACS pathophysiology. EXPERT OPINION Pathophysiology of ACS has been extensively investigated; however, several gray areas still remain. MINOCA represents one of these areas. At the same time, many aspects of immune-inflammation processes are still unknown. Thus, research should be continued to shed a brighter light on both these sides of "ACS" moon.
Collapse
Affiliation(s)
- Giovanni Cimmino
- Department of Translational Medical Sciences, Section of Cardiology, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Luigi Di Serafino
- Department of Advanced Biomedical Sciences, University of Naples Federico II, Naples, Italy
| | - Plinio Cirillo
- Department of Advanced Biomedical Sciences, University of Naples Federico II, Naples, Italy
| |
Collapse
|
574
|
Peering into the crystal ball to predict plaque rupture. J Clin Lipidol 2022; 16:383-385. [DOI: 10.1016/j.jacl.2022.05.066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Revised: 05/17/2022] [Accepted: 05/17/2022] [Indexed: 11/24/2022]
|
575
|
Statin Therapy for Primary Prevention in Women: What is the Role for Coronary Artery Calcium? J Clin Lipidol 2022; 16:376-382. [DOI: 10.1016/j.jacl.2022.05.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2022] [Revised: 04/28/2022] [Accepted: 05/03/2022] [Indexed: 12/30/2022]
|
576
|
Locatelli L, Castiglioni S, Maier JAM. From Cultured Vascular Cells to Vessels: The Cellular and Molecular Basis of Vascular Dysfunction in Space. Front Bioeng Biotechnol 2022; 10:862059. [PMID: 35480977 PMCID: PMC9036997 DOI: 10.3389/fbioe.2022.862059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Accepted: 03/18/2022] [Indexed: 11/23/2022] Open
Abstract
Life evolved on this planet under the pull of gravity, shielded from radiation by the magnetosphere and shaped by circadian rhythms due to Earth’s rotation on its axis. Once living beings leave such a protective environment, adaptive responses are activated to grant survival. In view of long manned mission out of Earth’s orbit, it is relevant to understand how humans adapt to space and if the responses activated might reveal detrimental in the long run. Here we review present knowledge about the effects on the vessels of various extraterrestrial factors on humans as well as in vivo and in vitro experimental models. It emerges that the vasculature activates complex adaptive responses finalized to supply oxygen and nutrients to all the tissues and to remove metabolic waste and carbon dioxide. Most studies point to oxidative stress and mitochondrial dysfunction as mediators of vascular alterations in space. Unraveling the cellular and molecular mechanisms involved in these adaptive processes might offer hints to design proper and personalized countermeasures to predict a safe future in space.
Collapse
Affiliation(s)
- Laura Locatelli
- Department of Biomedical and Clinical Sciences L. Sacco, Università di Milano, Milano, Italy
| | - Sara Castiglioni
- Department of Biomedical and Clinical Sciences L. Sacco, Università di Milano, Milano, Italy
| | - Jeanette A M Maier
- Department of Biomedical and Clinical Sciences L. Sacco, Università di Milano, Milano, Italy.,Interdisciplinary Centre for Nanostructured Materials and Interfaces (CIMaINa), Università di Milano, Milan, Italy
| |
Collapse
|
577
|
Baessler A, Fischer M. [Novel options to maximize oral lipid lowering treatment : Role of bempedoic acid in combination treatment]. Herz 2022; 47:212-219. [PMID: 35486131 DOI: 10.1007/s00059-022-05111-z] [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] [Accepted: 03/23/2022] [Indexed: 11/26/2022]
Abstract
Epidemiological, randomized, controlled, clinical and genetic studies confirm that low-density lipoprotein cholesterol (LDL-C) is a causative factor for atherosclerotic diseases. The current European Society of Cardiology and European Atherosclerosis Society (ESC/EAS) guidelines on the management of dyslipidemia recommend a target LDL-C < 55 mg/dl and at least a 50% reduction in baseline LDL‑C for high-risk patients; however, these target values are often not achieved in routine clinical practice, as shown by recent cross-sectional data from EUROASPIRE or DaVinci. Therefore, combination treatment is recommended, which, as with treatment of blood pressure, can improve the success of treatment. Bempedoic acid is a new substance, which is suitable for combination treatment and represents an alternative particularly for patients with statin-associated muscular symptoms. Bempedoic acid reduces LDL‑C by approximately 25% in statin-naïve patients and by some 18% in addition to statins. In a fixed doses combination with ezetimibe, bempedoic acid can lower LDL‑C by up to 45% in statin-naïve patients and by 38% (placebo-corrected) in addition to statins. Bempedoic acid is generally very well tolerated: however, it can lead to a reversible increase in uric acid. Occasionally, a slight decrease in hemoglobin has been documented. Therefore, it is recommended that not only changes in lipid levels but also uric acid and hematological parameters should be monitored in the first 3 months.
Collapse
Affiliation(s)
- Andrea Baessler
- Klinik und Poliklinik für Innere Medizin II, Universitätsklinikum Regensburg, Franz-Josef-Strauß-Allee 11, 93053, Regensburg, Deutschland.
| | - Marcus Fischer
- Klinik und Poliklinik für Innere Medizin II, Universitätsklinikum Regensburg, Franz-Josef-Strauß-Allee 11, 93053, Regensburg, Deutschland
- Klinik für Kardiologie, Angiologie, Pneumologie und Intensivmedizin, Goldberg-Klinik Kelheim, Kelheim, Deutschland
| |
Collapse
|
578
|
Quirós-Fernández R, López-Plaza B, Bermejo LM, Palma Milla S, Zangara A, Candela CG. Oral Supplement Containing Hydroxytyrosol and Punicalagin Improves Dyslipidemia in an Adult Population without Co-Adjuvant Treatment: A Randomized, Double-Blind, Controlled and Crossover Trial. Nutrients 2022; 14:nu14091879. [PMID: 35565844 PMCID: PMC9103949 DOI: 10.3390/nu14091879] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Revised: 04/22/2022] [Accepted: 04/26/2022] [Indexed: 12/04/2022] Open
Abstract
Hydroxytyrosol (HT) and punicalagin (PC) exert cardioprotective and antiatherosclerotic effects. This study evaluated the effect of an oral supplement containing HT and PC (SAx) on dyslipidemia in an adult population. A randomized, double-blind, controlled, crossover trial was conducted over a 20-week period. SAx significantly reduced the plasma levels of triglycerides (TG) in subjects with hypertriglyceridemia (≥150 mg/dL) (from 200.67 ± 51.38 to 155.33 ± 42.44 mg/dL; p < 0.05), while no such effects were observed in these subjects after the placebo. SAx also significantly decreased the plasma levels of low-density lipoprotein cholesterol (LDL-C) in subjects with high plasma levels of LDL-C (≥160 mg/dL) (from 179.13 ± 16.18 to 162.93 ± 27.05 mg/dL; p < 0.01), while no such positive effect was observed with the placebo. In addition, the placebo significantly reduced the plasma levels of high-density lipoprotein cholesterol (HDL-C) in the total population (from 64.49 ± 12.65 to 62.55 ± 11.57 mg/dL; p < 0.05), while SAx significantly increased the plasma levels of HDL-C in subjects with low plasma levels of HDL-C (<50 mg/dL) (from 44.25 ± 3.99 to 48.00 ± 7.27 mg/dL; p < 0.05). In conclusion, the supplement containing HT and PC exerted antiatherosclerotic and cardio-protective effects by considerably improving dyslipidemia in an adult population, without co-adjuvant treatment or adverse effects.
Collapse
Affiliation(s)
- Rebeca Quirós-Fernández
- Nutrition Research Group, Hospital La Paz Institute for Health Research (IdiPAZ), 28046 Madrid, Spain;
- Correspondence: (R.Q.-F.); (B.P.-L.)
| | - Bricia López-Plaza
- Nutrition Research Group, Hospital La Paz Institute for Health Research (IdiPAZ), 28046 Madrid, Spain;
- Correspondence: (R.Q.-F.); (B.P.-L.)
| | - Laura M. Bermejo
- Nutrition Research Group, Hospital La Paz Institute for Health Research (IdiPAZ), 28046 Madrid, Spain;
| | - Samara Palma Milla
- Nutrition Department, Hospital University La Paz, 28046 Madrid, Spain; (S.P.M.); (C.G.C.)
| | - Andrea Zangara
- Centre for Human Psychopharmacology, Swinburne University, Melbourne, VIC 3122, Australia;
- Euromed S.A., C/Rec de Dalt, 21-23, Pol. Ind. Can Magarola, 08100 Mollet del Valles, Spain
| | - Carmen Gómez Candela
- Nutrition Department, Hospital University La Paz, 28046 Madrid, Spain; (S.P.M.); (C.G.C.)
| |
Collapse
|
579
|
Li A, Li M, Guo J, Yun W, He Q. Efficacy of oats for dyslipidaemia: protocol for a systematic review and meta-analysis. BMJ Open 2022; 12:e058291. [PMID: 35487520 PMCID: PMC9052058 DOI: 10.1136/bmjopen-2021-058291] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Accepted: 03/01/2022] [Indexed: 11/23/2022] Open
Abstract
INTRODUCTION Dyslipidaemia is a critical factor in the development of atherosclerotic cardiovascular disease. Concerning dyslipidaemia regulation, we advocate for lifestyle interventions such as diet to complement drug treatment. Numerous studies have confirmed that oat β-glucan, a critical component of oats, can help lower cholesterol. However, there is no conclusive evidence for the efficacy of oats and their products in the treatment of dyslipidaemia. As a result, we have developed this protocol to serve as a guide for future research on oat intervention for dyslipidaemias. METHODS AND ANALYSIS We will conduct a search of eight databases or websites (PubMed, Web of Science, Cochrane Library, EMBASE, CNKI, SinoMed, VIP and Wanfang) to identify studies on oats' ability to regulate blood lipid levels. Two authors will screen articles independently, extract data based on inclusion and exclusion criteria, and assess the quality and bias of included studies. To assess and quantify heterogeneity, Q and I2 statistics will be used. If there is significant heterogeneity between studies, the source of the heterogeneity will be investigated using subgroup analysis and sensitivity analysis. We will analyse potential publication bias using the Begg funnel plot and Egger's weighted regression statistics. To assess the quality of evidence for the primary outcomes, the Grades of Recommendations, Assessment, Development and Evaluation method will be used. ETHICS AND DISSEMINATION This study is based on the existing literature and data in the databases. It is not subject to ethical review. The findings, on the other hand, will be published in a peer-reviewed journal. These findings may aid in the management of dyslipidaemia on a daily basis. PROSPERO REGISTRATION NUMBER CRD42021226751.
Collapse
Affiliation(s)
- Anqi Li
- Department of Cardiology, China Academy of Traditional Chinese Medicine Guanganmen Hospital, Beijing, China
- Beijing University of Traditional Chinese Medicine, Beijing, China
| | - Min Li
- Department of Cardiology, China Academy of Traditional Chinese Medicine Guanganmen Hospital, Beijing, China
| | - Jianbo Guo
- Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Wingyan Yun
- Department of Cardiology, China Academy of Traditional Chinese Medicine Guanganmen Hospital, Beijing, China
- Beijing University of Traditional Chinese Medicine, Beijing, China
| | - Qingyong He
- Department of Cardiology, China Academy of Traditional Chinese Medicine Guanganmen Hospital, Beijing, China
| |
Collapse
|
580
|
Kotlyarov S, Kotlyarova A. Molecular Pharmacology of Inflammation Resolution in Atherosclerosis. Int J Mol Sci 2022; 23:4808. [PMID: 35563200 PMCID: PMC9104781 DOI: 10.3390/ijms23094808] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 04/18/2022] [Accepted: 04/25/2022] [Indexed: 02/01/2023] Open
Abstract
Atherosclerosis is one of the most important problems of modern medicine as it is the leading cause of hospitalizations, disability, and mortality. The key role in the development and progression of atherosclerosis is the imbalance between the activation of inflammation in the vascular wall and the mechanisms of its control. The resolution of inflammation is the most important physiological mechanism that is impaired in atherosclerosis. The resolution of inflammation has complex, not fully known mechanisms, in which lipid mediators derived from polyunsaturated fatty acids (PUFAs) play an important role. Specialized pro-resolving mediators (SPMs) represent a group of substances that carry out inflammation resolution and may play an important role in the pathogenesis of atherosclerosis. SPMs include lipoxins, resolvins, maresins, and protectins, which are formed from PUFAs and regulate many processes related to the active resolution of inflammation. Given the physiological importance of these substances, studies examining the possibility of pharmacological effects on inflammation resolution are of interest.
Collapse
Affiliation(s)
- Stanislav Kotlyarov
- Department of Nursing, Ryazan State Medical University, 390026 Ryazan, Russia
| | - Anna Kotlyarova
- Department of Pharmacology and Pharmacy, Ryazan State Medical University, 390026 Ryazan, Russia;
| |
Collapse
|
581
|
Burger AL, Pogran E, Muthspiel M, Kaufmann CC, Jäger B, Huber K. New Treatment Targets and Innovative Lipid-Lowering Therapies in Very-High-Risk Patients with Cardiovascular Disease. Biomedicines 2022; 10:biomedicines10050970. [PMID: 35625707 PMCID: PMC9138506 DOI: 10.3390/biomedicines10050970] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Revised: 04/15/2022] [Accepted: 04/19/2022] [Indexed: 11/16/2022] Open
Abstract
The effective and fast reduction of circulating low-density lipoprotein cholesterol (LDL-C) is a cornerstone for secondary prevention of atherosclerotic disease progression. Despite the substantial lipid-lowering effects of the established treatment option with statins and ezetimibe, a significant proportion of very-high-risk patients with cardiovascular disease do not reach the recommended treatment goal of <55 mg/dL (<1.4 mmol/L). Novel lipid-lowering agents, including the proprotein convertase subtilisin/kexin type 9 (PCSK9) antibodies alirocumab and evolocumab, the small interfering ribonucleotide acid (si-RNA) inclisiran, as well as the recently approved bempedoic acid, now complete the current arsenal of LDL-C lowering agents. These innovative therapies have demonstrated promising results in clinical studies. Besides a strong reduction of LDL-C by use of highly effective agents, there is still discussion as to whether a very rapid achievement of the treatment goal should be a new strategic approach in lipid-lowering therapy. In this review, we summarize evidence for the lipid-modifying properties of these novel agents and their safety profiles, and discuss their potential pleiotropic effects beyond LDL-C reduction (if any) as well as their effects on clinical endpoints as cardiovascular mortality. In addition to a treatment strategy of “the lower, the better”, we also discuss the concept of “the earlier, the better”, which may also add to the early clinical benefit of large LDL-C reduction after an acute ischemic event.
Collapse
Affiliation(s)
- Achim Leo Burger
- 3rd Medical Department with Cardiology and Intensive Care Medicine, Clinic Ottakring (Wilhelminenhospital), Montleartstrasse 37, 1160 Vienna, Austria; (A.L.B.); (E.P.); (M.M.); (C.C.K.); (B.J.)
| | - Edita Pogran
- 3rd Medical Department with Cardiology and Intensive Care Medicine, Clinic Ottakring (Wilhelminenhospital), Montleartstrasse 37, 1160 Vienna, Austria; (A.L.B.); (E.P.); (M.M.); (C.C.K.); (B.J.)
| | - Marie Muthspiel
- 3rd Medical Department with Cardiology and Intensive Care Medicine, Clinic Ottakring (Wilhelminenhospital), Montleartstrasse 37, 1160 Vienna, Austria; (A.L.B.); (E.P.); (M.M.); (C.C.K.); (B.J.)
| | - Christoph Clemens Kaufmann
- 3rd Medical Department with Cardiology and Intensive Care Medicine, Clinic Ottakring (Wilhelminenhospital), Montleartstrasse 37, 1160 Vienna, Austria; (A.L.B.); (E.P.); (M.M.); (C.C.K.); (B.J.)
| | - Bernhard Jäger
- 3rd Medical Department with Cardiology and Intensive Care Medicine, Clinic Ottakring (Wilhelminenhospital), Montleartstrasse 37, 1160 Vienna, Austria; (A.L.B.); (E.P.); (M.M.); (C.C.K.); (B.J.)
| | - Kurt Huber
- 3rd Medical Department with Cardiology and Intensive Care Medicine, Clinic Ottakring (Wilhelminenhospital), Montleartstrasse 37, 1160 Vienna, Austria; (A.L.B.); (E.P.); (M.M.); (C.C.K.); (B.J.)
- Medical School, Sigmund Freud University, 1020 Vienna, Austria
- Correspondence: ; Tel.: +43-1-49150-2301
| |
Collapse
|
582
|
Karczewska-Kupczewska M, Nikołajuk A, Kondraciuk M, Stachurska Z, Dubatówka M, Szpakowicz A, Strączkowski M, Kowalska I, Kamiński K. The relationships between FLAIS, a novel insulin sensitivity index, and cardiovascular risk factors in a population-based study. Cardiovasc Diabetol 2022; 21:55. [PMID: 35439985 PMCID: PMC9020075 DOI: 10.1186/s12933-022-01491-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Accepted: 03/19/2022] [Indexed: 12/04/2022] Open
Abstract
Background Insulin resistance is a risk factor for cardiovascular disease. Recently, we have developed a novel index, FLAIS (Fasting Laboratory Assessment of Insulin Sensitivity), which accurately reflects insulin sensitivity, measured with hyperinsulinemic-euglycemic clamp, in different groups of subjects. The aim of the present study was to assess the relationship of FLAIS with cardiovascular risk factors in a population-based study. Methods The study group comprised 339 individuals from the ongoing Białystok Plus study, without previously known diabetes. Clinical examination, oral glucose tolerance test and the measurement of blood laboratory parameters were performed. Results Prediabetes (impaired fasting glucose and/or impaired glucose tolerance) was diagnosed in 165 individuals whereas type 2 diabetes was diagnosed in 19 subjects. FLAIS was lower in individuals with prediabetes and diabetes in comparison with individuals with normal glucose tolerance. FLAIS was significantly related to waist circumference, systolic and diastolic blood pressure, triglycerides, HDL-cholesterol and LDL-cholesterol in the entire study group and in the subgroups with normal glucose tolerance and with prediabetes/diabetes. HOMA-IR, QUICKI and Matsuda index were not related to blood pressure and LDL-cholesterol in individuals with normal glucose tolerance. Majority of the adjusted models with FLAIS were characterized by better fit with the data in comparison with other indices for all cardiovascular risk factors except waist circumference. Conclusions FLAIS represents useful index to assess the cluster of insulin resistance-associated cardiovascular risk factors in general population.
Collapse
Affiliation(s)
- Monika Karczewska-Kupczewska
- Department of Internal Medicine and Metabolic Diseases, Medical University of Białystok, M.C. Skłodowskiej 24a, 15-276, Białystok, Poland.
| | - Agnieszka Nikołajuk
- Department of Prophylaxis of Metabolic Diseases, Institute of Animal Reproduction and Food Research, Polish Academy of Sciences, Olsztyn, Poland
| | - Marcin Kondraciuk
- Department of Population Medicine and Lifestyle Diseases Prevention, Medical University of Białystok, Białystok, Poland
| | - Zofia Stachurska
- Department of Population Medicine and Lifestyle Diseases Prevention, Medical University of Białystok, Białystok, Poland
| | - Marlena Dubatówka
- Department of Population Medicine and Lifestyle Diseases Prevention, Medical University of Białystok, Białystok, Poland
| | - Anna Szpakowicz
- Department of Cardiology, Medical University of Białystok, Białystok, Poland
| | - Marek Strączkowski
- Department of Prophylaxis of Metabolic Diseases, Institute of Animal Reproduction and Food Research, Polish Academy of Sciences, Olsztyn, Poland
| | - Irina Kowalska
- Department of Internal Medicine and Metabolic Diseases, Medical University of Białystok, M.C. Skłodowskiej 24a, 15-276, Białystok, Poland
| | - Karol Kamiński
- Department of Population Medicine and Lifestyle Diseases Prevention, Medical University of Białystok, Białystok, Poland
| |
Collapse
|
583
|
Tan Y, Lin Q, Xu J, Zhu L, Guo L, Xie Y, Du X, Zhang S, Wen T, Liu L. Non-fasting Changes in Blood Lipids After Three Daily Meals Within a Day in Chinese Inpatients With Cardiovascular Diseases. Front Cardiovasc Med 2022; 9:799300. [PMID: 35498036 PMCID: PMC9039513 DOI: 10.3389/fcvm.2022.799300] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Accepted: 02/28/2022] [Indexed: 11/13/2022] Open
Abstract
Background Non-fasting (i.e., postprandial) lipid detection is recommended in clinical practice. However, the change in blood lipids in Chinese patients with cardiovascular diseases after three daily meals has never been reported yet. Methods Serum levels of blood lipids were measured or calculated in 77 inpatients (48 men and 29 women) at high or very high risk of atherosclerotic cardiovascular disease (ASCVD) in the fasting state and at 4 h after three meals within a day according to their diet habits. Results Female patients showed significantly higher level of high-density lipoprotein cholesterol (HDL-C) than male patients, and the gender difference in other lipid parameters did not reach statistical significance at any time-point. Levels of triglyceride (TG) and remnant cholesterol (RC) increased, while that of low-density lipoprotein cholesterol (LDL-C) decreased significantly after three meals (p < 0.05). Levels of HDL-C, total cholesterol (TC), and non-high-density lipoprotein cholesterol (non-HDL-C) showed smaller changes after three meals. Percent reductions in the non-fasting LDL-C levels after lunch and supper were around 20%, which were greater than that after breakfast. The percent reductions in the non-fasting non-HDL-C levels after three meals were smaller than those in the non-fasting LDL-C levels. Patients with TG level ≥ 2.0 mmol/L (177 mg/dL) after lunch had significantly greater absolute reduction of LDL-C level than those with TG level < 2.0 mmol/L (177 mg/dL) after lunch [–0.69 mmol/L (–27 mg/dL) vs. –0.36 mmol/L (–14 mg/dL), p<0.01]. There was a significant and negative correlation between absolute change in LDL-C level and that in TG level (r = −0.32) or RC level (r = −0.67) after lunch (both p<0.01). Conclusion LDL-C level decreased significantly after three daily meals in Chinese patients at high or very high risk of ASCVD, especially when TG level reached its peak after lunch. Relatively, non-HDL-C level was more stable than LDL-C level postprandially. Therefore, when LDL-C level was measured in the non-fasting state, non-HDL-C level could be evaluated simultaneously to reduce the interference of related factors, such as postprandial hypertriglyceridemia, on detection.
Collapse
Affiliation(s)
- Yangrong Tan
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha, China
- Research Institute of Blood Lipid and Atherosclerosis, Central South University, Changsha, China
- Modern Cardiovascular Disease Clinical Technology Research Center of Hunan Province, Changsha, China
- Cardiovascular Disease Research Center of Hunan Province, Changsha, China
| | - Qiuzhen Lin
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha, China
- Research Institute of Blood Lipid and Atherosclerosis, Central South University, Changsha, China
- Modern Cardiovascular Disease Clinical Technology Research Center of Hunan Province, Changsha, China
- Cardiovascular Disease Research Center of Hunan Province, Changsha, China
| | - Jin Xu
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha, China
- Research Institute of Blood Lipid and Atherosclerosis, Central South University, Changsha, China
- Modern Cardiovascular Disease Clinical Technology Research Center of Hunan Province, Changsha, China
- Cardiovascular Disease Research Center of Hunan Province, Changsha, China
| | - Liyuan Zhu
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha, China
- Research Institute of Blood Lipid and Atherosclerosis, Central South University, Changsha, China
- Modern Cardiovascular Disease Clinical Technology Research Center of Hunan Province, Changsha, China
- Cardiovascular Disease Research Center of Hunan Province, Changsha, China
| | - Liling Guo
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha, China
- Research Institute of Blood Lipid and Atherosclerosis, Central South University, Changsha, China
- Modern Cardiovascular Disease Clinical Technology Research Center of Hunan Province, Changsha, China
- Cardiovascular Disease Research Center of Hunan Province, Changsha, China
| | - Yingying Xie
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha, China
- Research Institute of Blood Lipid and Atherosclerosis, Central South University, Changsha, China
- Modern Cardiovascular Disease Clinical Technology Research Center of Hunan Province, Changsha, China
- Cardiovascular Disease Research Center of Hunan Province, Changsha, China
| | - Xiao Du
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha, China
- Research Institute of Blood Lipid and Atherosclerosis, Central South University, Changsha, China
- Modern Cardiovascular Disease Clinical Technology Research Center of Hunan Province, Changsha, China
- Cardiovascular Disease Research Center of Hunan Province, Changsha, China
| | - Shilan Zhang
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha, China
- Research Institute of Blood Lipid and Atherosclerosis, Central South University, Changsha, China
- Modern Cardiovascular Disease Clinical Technology Research Center of Hunan Province, Changsha, China
- Cardiovascular Disease Research Center of Hunan Province, Changsha, China
| | - Tie Wen
- Department of Emergency Medicine, The Second Xiangya Hospital, Central South University, Changsha, China
- Emergency Medicine and Difficult Diseases Institute, The Second Xiangya Hospital, Central South University, Changsha, China
- Tie Wen,
| | - Ling Liu
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha, China
- Research Institute of Blood Lipid and Atherosclerosis, Central South University, Changsha, China
- Modern Cardiovascular Disease Clinical Technology Research Center of Hunan Province, Changsha, China
- Cardiovascular Disease Research Center of Hunan Province, Changsha, China
- *Correspondence: Ling Liu,
| |
Collapse
|
584
|
Chazarin B, Benhaim-Delarbre M, Brun C, Anzeraey A, Bertile F, Terrien J. Molecular Liver Fingerprint Reflects the Seasonal Physiology of the Grey Mouse Lemur ( Microcebus murinus) during Winter. Int J Mol Sci 2022; 23:4254. [PMID: 35457071 PMCID: PMC9028843 DOI: 10.3390/ijms23084254] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Revised: 04/07/2022] [Accepted: 04/08/2022] [Indexed: 12/14/2022] Open
Abstract
Grey mouse lemurs (Microcebus murinus) are primates that respond to environmental energetic constraints through strong physiological seasonality. They notably fatten during early winter (EW), and mobilize their lipid reserves while developing glucose intolerance during late winter (LW), when food availability is low. To decipher how the hepatic mechanisms may support such metabolic flexibility, we analyzed the liver proteome of adult captive male mouse lemurs, whose seasonal regulations are comparable to their wild counterparts. We highlight profound hepatic changes that reflect fat accretion in EW at the whole-body level, without triggering an ectopic storage of fat in the liver, however. Moreover, molecular regulations are consistent with the decrease in liver glucose utilization in LW, and therefore with reduced tolerance to glucose. However, no major regulation was seen in insulin signaling/resistance pathways. Fat mobilization in LW appeared possibly linked to the reactivation of the reproductive system while enhanced liver detoxification may reflect an anticipation to return to summer levels of food intake. Overall, these results show that the physiology of mouse lemurs during winter relies on solid molecular foundations in liver processes to adapt fuel partitioning while opposing the development of a pathological state despite large lipid fluxes.
Collapse
Affiliation(s)
- Blandine Chazarin
- Laboratoire de Spectrométrie de Masse Bio-Organique, Institut Pluridisciplinaire Hubert Curien, University of Strasbourg, CNRS, UMR 7178, 25 Rue Becquerel, 67087 Strasbourg, France; (B.C.); (M.B.-D.); (C.B.)
- Infrastructure Nationale de Protéomique ProFI—FR 2048, 25 Rue Becquerel, 67087 Strasbourg, France
| | - Margaux Benhaim-Delarbre
- Laboratoire de Spectrométrie de Masse Bio-Organique, Institut Pluridisciplinaire Hubert Curien, University of Strasbourg, CNRS, UMR 7178, 25 Rue Becquerel, 67087 Strasbourg, France; (B.C.); (M.B.-D.); (C.B.)
- Infrastructure Nationale de Protéomique ProFI—FR 2048, 25 Rue Becquerel, 67087 Strasbourg, France
| | - Charlotte Brun
- Laboratoire de Spectrométrie de Masse Bio-Organique, Institut Pluridisciplinaire Hubert Curien, University of Strasbourg, CNRS, UMR 7178, 25 Rue Becquerel, 67087 Strasbourg, France; (B.C.); (M.B.-D.); (C.B.)
- Infrastructure Nationale de Protéomique ProFI—FR 2048, 25 Rue Becquerel, 67087 Strasbourg, France
| | - Aude Anzeraey
- Unité Mécanismes Adaptatifs et Evolution (MECADEV), UMR 7179, CNRS, Muséum National d’Histoire Naturelle, 1 Avenue du Petit Château, 91800 Brunoy, France;
| | - Fabrice Bertile
- Laboratoire de Spectrométrie de Masse Bio-Organique, Institut Pluridisciplinaire Hubert Curien, University of Strasbourg, CNRS, UMR 7178, 25 Rue Becquerel, 67087 Strasbourg, France; (B.C.); (M.B.-D.); (C.B.)
- Infrastructure Nationale de Protéomique ProFI—FR 2048, 25 Rue Becquerel, 67087 Strasbourg, France
| | - Jérémy Terrien
- Unité Mécanismes Adaptatifs et Evolution (MECADEV), UMR 7179, CNRS, Muséum National d’Histoire Naturelle, 1 Avenue du Petit Château, 91800 Brunoy, France;
| |
Collapse
|
585
|
Ye W, Tang Q, Wang L, Fang C, Xie L, He Q, Peng K. Contribution of CYP19A1, CYP1A1, and CYP1A2 polymorphisms in coronary heart disease risk among the Chinese Han population. Funct Integr Genomics 2022; 22:515-524. [PMID: 35380334 DOI: 10.1007/s10142-022-00850-y] [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: 09/09/2021] [Revised: 03/09/2022] [Accepted: 03/22/2022] [Indexed: 11/29/2022]
Abstract
The previous study has pointed to that endogenous CYP metabolites play an important role in the pathogenesis of coronary heart disease (CHD). The study aimed to identify the association of CYP19A1, CYP1A1, and CYP1A2 polymorphisms with CHD susceptibility in a Chinese Han population. A total of 960 genetically unrelated participants consist of 480 CHD patients and 480 healthy controls were enrolled. Nine SNPs in CYP19A1, CYP1A1, and CYP1A2 were randomly selected and genotyped using the Agena MassARRAY platform. Logistic regression analysis was used for the relationship between selected SNPs and CHD susceptibility by calculating odds ratios (OR) with 95% confidence intervals (CI) adjusted for age and gender. The distribution of clinical characteristics in different genotypes was evaluated by one-way analysis of variance (ANOVA). CYP1A2 rs2470890 TT genotype had a higher CHD risk compared with CC genotype (OR = 3.06, p = 0.032) or CC-CT genotype (OR = 3.04, p = 0.033). Moreover, the contribution of CYP19A1 and CYP1A2 polymorphisms to CHD susceptibility was associated with age, gender, and clinical phenotypes (course of the disease and Gensini score). Besides, CYP1A2 rs762551 was related to serum levels of red blood cell, triglyceride, total cholesterol, and low-density lipoprotein cholesterol (LDL-C, p < 0.05). Our findings provided scientific evidence about CYP19A1, CYP1A1, and CYP1A2 polymorphisms on CHD incidence.
Collapse
Affiliation(s)
- Wei Ye
- Department of Neonatology, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei Province, People's Republic of China.,Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei Province, People's Republic of China.,Department of Cardiology, Renmin Hospital of Wuhan University, # 99 Zhizhidong Road, Wuhan, Hubei, 430060, People's Republic of China.,Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, 430060, People's Republic of China
| | - Qizhu Tang
- Department of Cardiology, Renmin Hospital of Wuhan University, # 99 Zhizhidong Road, Wuhan, Hubei, 430060, People's Republic of China. .,Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, 430060, People's Republic of China.
| | - Lei Wang
- Department of Cardiology, HanChuan Hospital, Renmin Hospital of Wuhan University, Hanchuan, Hubei, 431600, People's Republic of China
| | - Chenzhi Fang
- Department of Neonatology, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei Province, People's Republic of China
| | - Lili Xie
- Department of Neonatology, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei Province, People's Republic of China
| | - Qi He
- Department of Neonatology, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei Province, People's Republic of China
| | - Kaiwei Peng
- Department of Neonatology, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei Province, People's Republic of China
| |
Collapse
|
586
|
Altschmiedova T, Todorovova V, Vrablik M, Ceska R. Familial Hypercholesterolemia: Real-World Data of 1236 Patients Attending a Czech Lipid Clinic. A Retrospective Analysis of Experience in More than 50 years. Part II. Clinical Characteristics. Front Genet 2022; 13:849267. [PMID: 35368707 PMCID: PMC8964355 DOI: 10.3389/fgene.2022.849267] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Accepted: 02/02/2022] [Indexed: 11/13/2022] Open
Abstract
Introduction: Patients with familial hypercholesterolemia (FH) are at increased risk of premature atherosclerotic cardiovascular disease (ASCVD). Aim of study: To perform a retrospective analysis of data to assess the effects of individual lipoproteins and other risk factors (RFs) on the development of ASCVD and to compare these parameters in individuals with versus without ASCVD. Patients and methods: Our study group included a total of 1,236 patients with FH (395 men and 841 women with a mean age of 44.8 ± 16.7 years) attending a single lipid clinic. The diagnosis of FH was established using the Dutch Lipid Clinic Network score (DLCN). Among the 1236 FH patients, 1,008 of them [854 suspected with LDL receptor-mediated FH and 154 with familial defective apolipoprotein B-100 (FDB)] were genetically analysed. Their RFs were assessed based on the patients’ clinical characteristics. Results: While patients with ASCVD had higher baseline LDL-C, TC, TG and Lp(a) compared with patients without this diagnosis, this ratio was just the opposite by the follow-up. The highest statistically significant differences were seen in the baseline levels of Lp(a) and, quite surprisingly, TG. Except for Lp(a), the levels of all lipid parameters declined significantly over time. While the incidence of diabetes and arterial hypertension was not higher in our group compared with the general population, these patients were at a more significant risk of ASCVD. Conclusion: Familial hypercholesterolemia is a major RF for the development of ASCVD. While our analysis confirmed the important role of LDL-C, it also corroborated a strong correlation between ASCVD and other lipid parameters, and Lp(a) and TG in particular. Familial hypercholesterolemia is not the only RF and, to reduce cardiovascular risk of their patients, physicians have to search for other potential RFs. Patients diagnosed to have FH benefit from attending a specialized lipid clinic perse.
Collapse
Affiliation(s)
- Tereza Altschmiedova
- Third Department of Medicine - Department of Endocrinology and Metabolism of the First Faculty of Medicine, Charles University and General University Hospital, Prague, Czechia
| | - Veronika Todorovova
- Third Department of Medicine - Department of Endocrinology and Metabolism of the First Faculty of Medicine, Charles University and General University Hospital, Prague, Czechia
| | - Michal Vrablik
- Third Department of Medicine - Department of Endocrinology and Metabolism of the First Faculty of Medicine, Charles University and General University Hospital, Prague, Czechia
| | - Richard Ceska
- Third Department of Medicine - Department of Endocrinology and Metabolism of the First Faculty of Medicine, Charles University and General University Hospital, Prague, Czechia
| |
Collapse
|
587
|
Cirillo P, Conte S, Pellegrino G, Barra G, De Palma R, Sugraliyev A, Golino P, Cimmino G. Effects of colchicine on tissue factor in oxLDL-activated T-lymphocytes. J Thromb Thrombolysis 2022; 53:739-749. [PMID: 34671897 DOI: 10.1007/s11239-021-02585-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/30/2021] [Indexed: 12/17/2022]
Abstract
Several studies have shown that T-cells might be involved in pathophysiology of acute coronary syndromes (ACS). Tissue factor (TF) plays a key role in ACS. Many evidences have indicated that some statins reduce TF expression in several cell types. However, literature about rosuvastatin and TF and about statins effects on T-cells is still scanty. Colchicine is an anti-inflammatory drug recently proven to have beneficial effects in ACS via unknown mechanisms. This study investigates the effects of colchicine and rosuvastatin on TF expression in oxLDL-activated T-cells. T-cells, isolated from buffy coats of healthy volunteers, were stimulated with oxLDL (50 µg/dL). T-cells were pre-incubated with colchicine (10 µM) or rosuvastatin (5 µM) for 1 h and then stimulated with oxLDL (50 μg/mL). TF gene (RT-PCR), protein (western blot), surface expression (FACS) and procoagulant activity (FXa generation assay) were measured. NF-κB/IκB axis was examined by western blot analysis and translocation assay. Colchicine and rosuvastatin significantly reduced TF gene, and protein expression and procoagulant activity in oxLDL stimulated T-cells. This effect was associated with a significant reduction in TF surface expression as well as its procoagulant activity. These phenomena appear modulated by drug effects on the transcription factor NF-kB. Rosuvastatin and colchicine prevent TF expression in oxLDL-stimulated T-cells by modulating the NF-κB/IκB axis. Thus, we speculate that this might be another mechanism by which these drugs exert benefic cardiovascular effects.
Collapse
Affiliation(s)
- Plinio Cirillo
- Division of Cardiology, Department of Advanced Biomedical Sciences, University of Naples "Federico II", Via Sergio Pansini 5, 80131, Naples, Italy.
| | - Stefano Conte
- Department of Translational Medical Sciences, Section of Cardiology, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Grazia Pellegrino
- Division of Cardiology, Department of Advanced Biomedical Sciences, University of Naples "Federico II", Via Sergio Pansini 5, 80131, Naples, Italy
| | - Giusi Barra
- Unit of Internal Medicine, Clinical Immunology and Translational Medicine, IRCCS Ospedale Policlinico San Martino, University of Genova, Genova, Italy
| | - Raffaele De Palma
- Unit of Internal Medicine, Clinical Immunology and Translational Medicine, IRCCS Ospedale Policlinico San Martino, University of Genova, Genova, Italy
| | - Akhmetzhan Sugraliyev
- Department of Internal Disease, Kazakh National Medical University, Almaty, Kazakhstan
| | - Paolo Golino
- Department of Translational Medical Sciences, Section of Cardiology, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Giovanni Cimmino
- Department of Translational Medical Sciences, Section of Cardiology, University of Campania "Luigi Vanvitelli", Naples, Italy
| |
Collapse
|
588
|
Lucci C, van den Beukel TC, Bartstra JW, Zwanenburg J, van der Kolk A, Takx R, Hendrikse J, Geerlings MI, Bos D, Spiering W, de Jong PA. Intracranial atherosclerosis in pseudoxanthoma elasticum: A case-control study. Atherosclerosis 2022; 350:19-24. [DOI: 10.1016/j.atherosclerosis.2022.04.014] [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: 12/20/2021] [Revised: 04/08/2022] [Accepted: 04/13/2022] [Indexed: 11/25/2022]
|
589
|
Jebari-Benslaiman S, Uribe KB, Benito-Vicente A, Galicia-Garcia U, Larrea-Sebal A, Santin I, Alloza I, Vandenbroeck K, Ostolaza H, Martín C. Boosting Cholesterol Efflux from Foam Cells by Sequential Administration of rHDL to Deliver MicroRNA and to Remove Cholesterol in a Triple-Cell 2D Atherosclerosis Model. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2022; 18:e2105915. [PMID: 35156292 DOI: 10.1002/smll.202105915] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Revised: 12/30/2021] [Indexed: 06/14/2023]
Abstract
Cardiovascular disease, the leading cause of mortality worldwide, is primarily caused by atherosclerosis, which is characterized by lipid and inflammatory cell accumulation in blood vessels and carotid intima thickening. Although disease management has improved significantly, new therapeutic strategies focused on accelerating atherosclerosis regression must be developed. Atherosclerosis models mimicking in vivo-like conditions provide essential information for research and new advances toward clinical application. New nanotechnology-based therapeutic opportunities have emerged with apoA-I nanoparticles (recombinant/reconstituted high-density lipoproteins, rHDL) as ideal carriers to deliver molecules and the discovery that microRNAs participate in atherosclerosis establishment and progression. Here, a therapeutic strategy to improve cholesterol efflux is developed based on a two-step administration of rHDL consisting of a first dose of antagomiR-33a-loaded rHDLs to induce adenosine triphosphate-binding cassette transporters A1 overexpression, followed by a second dose of 1,2-dipalmitoyl-sn-glycero-3-phosphocholine rHDLs, which efficiently remove cholesterol from foam cells. A triple-cell 2D-atheroma plaque model reflecting the cellular complexity of atherosclerosis is used to improve efficiency of the nanoparticles in promoting cholesterol efflux. The results show that sequential administration of rHDL potentiates cholesterol efflux indicating that this approach may be used in vivo to more efficiently target atherosclerotic lesions and improve prognosis of the disease.
Collapse
Affiliation(s)
- Shifa Jebari-Benslaiman
- Biofisika Institute (UPV/EHU, CSIC) and Department of Biochemistry and Molecular Biology, University of the Basque Country UPV/EHU, Leioa, 48940, Spain
| | - Kepa B Uribe
- Center for Cooperative Research in Biomaterials (CIC biomaGUNE), Basque Research and Technology Alliance (BRTA), San Sebastián, 20014, Spain
| | - Asier Benito-Vicente
- Biofisika Institute (UPV/EHU, CSIC) and Department of Biochemistry and Molecular Biology, University of the Basque Country UPV/EHU, Leioa, 48940, Spain
| | - Unai Galicia-Garcia
- Fundación Biofisika Bizkaia and Biofisika Institute (UPV/EHU, CSIC), Leioa, 48940, Spain
| | - Asier Larrea-Sebal
- Fundación Biofisika Bizkaia and Biofisika Institute (UPV/EHU, CSIC), Leioa, 48940, Spain
| | - Izortze Santin
- Department of Biochemistry and Molecular biology, University of the Basque Country UPV/EHU, Leioa, 48940, Spain
- Biocruces Bizkaia Health Research Institute, Barakaldo, 48903, Spain
- CIBER (Centro de Investigación Biomédica en Red) de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III, Spain
| | - Iraide Alloza
- Biocruces Bizkaia Health Research Institute, Barakaldo, 48903, Spain
| | - Koen Vandenbroeck
- Biocruces Bizkaia Health Research Institute, Barakaldo, 48903, Spain
| | - Helena Ostolaza
- Biofisika Institute (UPV/EHU, CSIC) and Department of Biochemistry and Molecular Biology, University of the Basque Country UPV/EHU, Leioa, 48940, Spain
| | - César Martín
- Biofisika Institute (UPV/EHU, CSIC) and Department of Biochemistry and Molecular Biology, University of the Basque Country UPV/EHU, Leioa, 48940, Spain
| |
Collapse
|
590
|
Bilitou A, Were J, Farrer A, Rabe A, Ming SWY, Haq I, Dunton K. Prevalence and Patient Outcomes of Adult Primary Hypercholesterolemia and Dyslipidemia in the UK: Longitudinal Retrospective Study Using a Primary Care Dataset from 2009 to 2019. CLINICOECONOMICS AND OUTCOMES RESEARCH 2022; 14:189-203. [PMID: 35411162 PMCID: PMC8994561 DOI: 10.2147/ceor.s347085] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Accepted: 03/10/2022] [Indexed: 01/06/2023] Open
Abstract
Background Guidelines for the management of dyslipidemias recommend intensive low-density lipoprotein (LDL-C) control through lifestyle advice and lipid-lowering drugs to reduce the risk of cardiovascular disease (CVD). Objective This retrospective study aimed to characterize the adult primary care population with primary hypercholesterolemia (PH)/mixed dyslipidemia (MD). Methods Data on adults with PH/MD between 1 January 2009 and 31 December 2019 in the UK were extracted from linked primary Clinical Practice Research Datalink (CPRD) and secondary care (Hospital Episode Statistics) datasets and analyzed. Results A total of 279,221 patients met the inclusion criteria. Mean follow-up was 8.6 years. Crude prevalence of PH/MD increased from 13.5% in 2009 to 23.5% by 2019. The incidence decreased from 176 to 49 per 100,000 population. Mean age of the cohort was 58 years, baseline LDL-C was 4.32 mmol/L, 19.6% had atherosclerotic CVD, 30.1% diabetes, and 8.5% heterozygous familial hypercholesterolemia. Estimated LDL-C reductions of 40% and 50% were achieved in 2.6% and 2.3% of patients, respectively. Most received moderate-intensity statins as monotherapy (62.4%); high-intensity statins were used less frequently (24.3% as initial treatment). Less than 10% of patients received ezetimibe plus statins of different intensities. Conclusion The prevalence of dyslipidemia doubled between 2009 and 2019, likely due to more systematic identification of PH/MD. A large proportion of patients with PH/MD are of high and very high CV risk, remain suboptimally treated in terms of lipid lowering, and may experience CV events with associated non-negligible clinical and economic sequelae. Despite intensive LDL-C-lowering recommendations, these do not translate in clinical practice to the wider population.
Collapse
Affiliation(s)
- Aikaterini Bilitou
- Daiichi Sankyo Europe GmbH, Munich, Bayern, Germany
- Correspondence: Aikaterini Bilitou, Health Economics and Outcomes Research, Daiichi Sankyo Europe GmbH, Zielstattstrasse 48, Munich, 81379, Bayern, Germany, Tel +49 897 808802, Email
| | | | | | - Adrian Rabe
- Health iQ Ltd, London, UK
- Imperial College London, London, UK
| | | | - Inaam Haq
- Daiichi Sankyo Europe GmbH, Munich, Bayern, Germany
| | - Kyle Dunton
- Daiichi Sankyo UK Ltd, Uxbridge Business Park, Uxbridge, UK
| |
Collapse
|
591
|
Chary A, Hedayati M. Review of Laboratory Methods to Determine HDL and LDL Subclasses and Their Clinical Importance. Rev Cardiovasc Med 2022; 23:147. [PMID: 39076233 PMCID: PMC11273998 DOI: 10.31083/j.rcm2304147] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Revised: 02/01/2022] [Accepted: 02/15/2022] [Indexed: 07/31/2024] Open
Abstract
Given the high prevalence of cardiovascular disease, accurate identification of methods for assessing lipoprotein subclasses, mainly low-density lipoprotein (LDL) and high-density lipoprotein (HDL) subfractions, can play an essential role in predicting the incidence of cardiovascular disease such as heart attack. LDL and HDL subclasses differ in size, surface charge, lipid and protein compositions, and biological role. There is no "gold standard" method for measuring the LDL and HDL subclasses or standardizing the different methods used to measure their subfractions. Over the past decades, various techniques have been introduced to evaluate and measure subclasses of these two lipoproteins, each with its own advantages and disadvantages. Development of laboratory methods that accurately HDL and LDL function must be developed and validated to high-throughput for clinical usage. In this review study, we tried to examine different methods of evaluating various subclasses of LDL and HDL by mentioning the strengths and weaknesses of each.
Collapse
Affiliation(s)
- Abdolreza Chary
- Prevention of Metabolic Disorders Research Center, Research Institute for Endocrine Science, Shaheed Beheshti University of Medical Sciences, 1985717413 Tehran, Iran
| | - Mehdi Hedayati
- Cellular and Molecular Endocrine Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, 1985717413 Tehran, Iran
| |
Collapse
|
592
|
Fuller J. Preventive and Curative Medical Interventions. SYNTHESE 2022; 200:61. [PMID: 36090528 PMCID: PMC9455911 DOI: 10.1007/s11229-022-03579-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Accepted: 01/07/2022] [Indexed: 06/15/2023]
Abstract
Medical interventions that cure or prevent medical conditions are central to medicine; and thus, understanding them is central to our understanding of medicine. My purpose in this paper is to explore the conceptual foundations of medicine by providing a singular analysis of the concept of a 'preventive or curative medical intervention'. Borrowing a general account of prevention from Phil Dowe (2000, 2001), I provide an analysis of prevention, cure, risk reduction, and a preventive or curative intervention, before turning to preventive and curative medical interventions. The resulting counterfactual-mechanistic account holds that preventive and curative medical interventions reduce the probability of a medical condition in an actual population compared to their counterfactual omission, commonly by disrupting an etiological or constitutive mechanism for the condition.
Collapse
Affiliation(s)
- Jonathan Fuller
- Department of History and Philosophy of Science, University of Pittsburgh, Pittsburgh, PA, USA
| |
Collapse
|
593
|
Pasterkamp G, den Ruijter HM, Giannarelli C. False Utopia of One Unifying Description of the Vulnerable Atherosclerotic Plaque: A Call for Recalibration That Appreciates the Diversity of Mechanisms Leading to Atherosclerotic Disease. Arterioscler Thromb Vasc Biol 2022; 42:e86-e95. [PMID: 35139657 DOI: 10.1161/atvbaha.121.316693] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Atherosclerosis is a complex disease characterized by the formation of arterial plaques with a broad diversity of morphological phenotypic presentations. Researchers often apply one description of the vulnerable plaque as a gold standard in preclinical and clinical research that could be applied as a surrogate measure of a successful therapeutic intervention, despite the variability in lesion characteristics that may underly a thrombotic occlusion. The complex mechanistic interplay underlying progression of atherosclerotic disease is a consequence of the broad range of determinants such as sex, risk factors, hemodynamics, medications, and the genetic landscape. Currently, we are facing an overwhelming amount of data based on genetic, transcriptomic, proteomic, and metabolomic studies that all point to heterogeneous molecular profiles of atherosclerotic lesions that lead to a myocardial infarction or stroke. The observed molecular diversity implies that one unifying model cannot fully recapitulate the natural history of atherosclerosis. Despite emerging data obtained from -omics studies, a description of a natural history of atherosclerotic disease in which cell-specific expression of proteins or genes are included is still lacking. This also applies to the insights provided by genome-wide association studies. This review will critically discuss the dogma that the progression of atherosclerotic disease can be captured in one unifying natural history model of atherosclerosis.
Collapse
Affiliation(s)
- Gerard Pasterkamp
- Circulatory Health Laboratories (G.P., H.M.d.R.), University Medical Center Utrecht, the Netherlands.,Central Diagnostics Laboratories (G.P.), University Medical Center Utrecht, the Netherlands
| | - Hester M den Ruijter
- Circulatory Health Laboratories (G.P., H.M.d.R.), University Medical Center Utrecht, the Netherlands.,Laboratory of Experimental Cardiology (H.M.d.R.), University Medical Center Utrecht, the Netherlands
| | - Chiara Giannarelli
- NYU Cardiovascular Research Center (C.G.), New York University Grossman School of Medicine.,Department of Pathology (C.G.), New York University Grossman School of Medicine
| |
Collapse
|
594
|
Peanut skin extract ameliorates high-fat diet-induced atherosclerosis by regulating lipid metabolism, inflammation reaction and gut microbiota in ApoE−/− mice. Food Res Int 2022; 154:111014. [DOI: 10.1016/j.foodres.2022.111014] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Revised: 02/11/2022] [Accepted: 02/12/2022] [Indexed: 11/18/2022]
|
595
|
Limpijankit T, Vathesatogkit P, Matchariyakul D, Wiriyatanakorn S, Siriyotha S, Thakkinstian A, Sritara P. Causal relationship of excess body weight on cardiovascular events through risk factors. Sci Rep 2022; 12:5269. [PMID: 35347154 PMCID: PMC8960828 DOI: 10.1038/s41598-022-08812-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Accepted: 03/09/2022] [Indexed: 11/09/2022] Open
Abstract
Excess body weight is associated with cardiovascular events (CVEs) and premature death. This study aimed to find the causal pathways between excess body weight and CVEs through risk factors in a general adult population. A total of 7921 employees of the Electricity Generating Authority of Thailand were enrolled during 1997-2009. Baseline characteristics and blood test results were collected. A body mass index (BMI) ≥ 23 kg/m2, using WHO criteria for Asians was defined as excess body weight. A mediation analysis was applied to assess potential causal pathways. BMI ≥ 23 kg/m2 was considered as an independent variable, whereas diabetes mellitus (DM), hypertension (HT), and chronic kidney disease (CKD) were considered as mediators, and CVEs (i.e., fatal and non-fatal coronary artery disease or stroke) were considered as the outcomes. The prevalence of BMI ≥ 23 kg/m2, DM, HT, and CKD were 62.7%, 7.8%, 28.1% and 11.8% respectively. During an average of 17.2 ± 5.5 years follow-up, subjects with BMI ≥ 23 kg/m2 compared with those with lower BMIs more frequently developed CVEs (9.4 vs 6.2%, P < 0.001). The effects of BMI ≥ 23 kg/m2 on CVEs were mediated indirectly through DM and HT with significant ORs of 1.61 (1.34, 2.09) and 1.57 (1.39, 1.80), respectively. The indirect effect of CKD on CVEs was significantly increased if mediated through DM → HT or HT [ORs of 1.17 (1.09, 1.32) and 1.20 (1.10, 1.32), respectively]. Subjects with excess body weight were prone to develop CVEs which were mediated indirectly through DM and HT. The effect of CKD on CVEs was small but enhanced if it occurred as a complication of DM or HT.
Collapse
Affiliation(s)
- Thosaphol Limpijankit
- Division of Cardiology, Department of Medicine, Faculty of Medicine Ramathibodi Hospital, Mahidol University, 270 Rama VI Road, Ratchathewi, Bangkok, 10400, Thailand.
| | - Prin Vathesatogkit
- Division of Cardiology, Department of Medicine, Faculty of Medicine Ramathibodi Hospital, Mahidol University, 270 Rama VI Road, Ratchathewi, Bangkok, 10400, Thailand
| | - Dujrudee Matchariyakul
- Medical and Health Office, Electricity Generating Authority of Thailand, Bangkruay, Nonthaburi, 11130, Thailand
| | - Sirichai Wiriyatanakorn
- Division of Cardiology, Department of Medicine, Faculty of Medicine Ramathibodi Hospital, Mahidol University, 270 Rama VI Road, Ratchathewi, Bangkok, 10400, Thailand
| | - Sukanya Siriyotha
- Section for Clinical Epidemiology and Biostatistics, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - Ammarin Thakkinstian
- Section for Clinical Epidemiology and Biostatistics, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - Piyamitr Sritara
- Division of Cardiology, Department of Medicine, Faculty of Medicine Ramathibodi Hospital, Mahidol University, 270 Rama VI Road, Ratchathewi, Bangkok, 10400, Thailand
| |
Collapse
|
596
|
Arnaboldi L, Corsini A, Bellosta S. Artichoke and bergamot extracts: a new opportunity for the management of dyslipidemia and related risk factors. Minerva Med 2022; 113:141-157. [PMID: 35313442 DOI: 10.23736/s0026-4806.21.07950-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
The relationship between low LDL-C (cholesterol associated with low-density lipoprotein) and a lower relative risk of developing cardiovascular disease (CVD) has been widely demonstrated. Although from a pharmacological point of view, statins, ezetimibe and PCSK inhibitors, alone or in combination are the front and center of the therapeutic approaches for reducing LDL-C and its CV consequences, in recent years nutraceuticals and functional foods have increasingly been considered as a valid support in the reduction of LDL-C, especially in patients with mild/moderate hyperlipidemia - therefore not requiring pharmacological treatment - or in patients intolerant to statins or other drugs. An approach also shared by the European Atherosclerosis Society (EAS). Of the various active ingredients with hypolipidemic properties, we include the artichoke (Cynara cardunculus, Cynara scolymus) and the bergamot (Citrus bergamia) which, thanks essentially to the significant presence of polyphenols in their extracts, can exert this action associated with a number of other complementary inflammation and oxidation benefits. In light of these evidence, this review aimed to describe the effects of artichoke and bergamot in modifying the lipid and inflammatory parameters described in in vitro, in vivo and clinical studies. The available data support the use of standardized compositions of artichoke and bergamot extracts, alone or in combination, in the treatment of mild to moderate dyslipidemia, in patients suffering from metabolic syndrome, hepatic steatosis, or intolerant to common hypolipidemic treatments.
Collapse
|
597
|
Borén J, Taskinen MR, Björnson E, Packard CJ. Metabolism of triglyceride-rich lipoproteins in health and dyslipidaemia. Nat Rev Cardiol 2022; 19:577-592. [PMID: 35318466 DOI: 10.1038/s41569-022-00676-y] [Citation(s) in RCA: 118] [Impact Index Per Article: 39.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 02/02/2022] [Indexed: 02/07/2023]
Abstract
Accumulating evidence points to the causal role of triglyceride-rich lipoproteins and their cholesterol-enriched remnants in atherogenesis. Genetic studies in particular have not only revealed a relationship between plasma triglyceride levels and the risk of atherosclerotic cardiovascular disease, but have also identified key proteins responsible for the regulation of triglyceride transport. Kinetic studies in humans using stable isotope tracers have been especially useful in delineating the function of these proteins and revealing the hitherto unappreciated complexity of triglyceride-rich lipoprotein metabolism. Given that triglyceride is an essential energy source for mammals, triglyceride transport is regulated by numerous mechanisms that balance availability with the energy demands of the body. Ongoing investigations are focused on determining the consequences of dysregulation as a result of either dietary imprudence or genetic variation that increases the risk of atherosclerosis and pancreatitis. The identification of molecular control mechanisms involved in triglyceride metabolism has laid the groundwork for a 'precision-medicine' approach to therapy. Novel pharmacological agents under development have specific molecular targets within a regulatory framework, and their deployment heralds a new era in lipid-lowering-mediated prevention of disease. In this Review, we outline what is known about the dysregulation of triglyceride transport in human hypertriglyceridaemia.
Collapse
Affiliation(s)
- Jan Borén
- Department of Molecular and Clinical Medicine, University of Gothenburg, Gothenburg, Sweden.
| | - Marja-Riitta Taskinen
- Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Elias Björnson
- Department of Molecular and Clinical Medicine, University of Gothenburg, Gothenburg, Sweden
| | - Chris J Packard
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, UK
| |
Collapse
|
598
|
Li Z, Tang Z, Wang Y, Liu Z, Wang G, Zhang L, Wu Y, Guo J. Assessment of radial artery atherosclerosis in acute coronary syndrome patients: an in vivo study using optical coherence tomography. BMC Cardiovasc Disord 2022; 22:120. [PMID: 35313827 PMCID: PMC8939080 DOI: 10.1186/s12872-022-02561-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Accepted: 03/14/2022] [Indexed: 11/23/2022] Open
Abstract
Background Radial artery (RA) atherosclerosis in acute coronary syndrome (ACS) patients has not been systematically observed in vivo. The study aims to characterize plaque morphology and intimal hyperplasia of the RA in patients with ACS, using optical coherence tomography (OCT). Methods In this retrospective study involving 239 ACS patients underwent RA OCT without guidewire shadow, 3 groups were divided according to the following criteria: radial artery plaque (RAP) group included patients with fibrous, lipid or calcified plaque; patients without RAP were further classified into radial intimal hyperplasia (RIH) group (intima media thickness ratio [IMR] ≥ 1) or normal group (IMR < 1). The presence and characteristics of RAP and its related risk factors were identified. Results The RAP, RIH and normal groups included 76 (31.8%), 69 (28.9%) and 94 (39.3%) patients, respectively. Patients in RAP group were the oldest, compared with those in the RIH and normal groups (p < 0.001), and more frequently had triple vessel disease (p = 0.004). The percentage of plaque rupture (72.4% vs. 56.4%, p = 0.018) and calcification (42.1% vs. 27.6%, p = 0.026) at culprit lesion were significantly higher in patients with RAP than those without RAP. A total of 148 RAP were revealed by OCT, including fibrous (72, 48.6%), lipid (50, 33.8%) and calcified plaques (26, 17.6%). The microvessels were also frequently observed in the RAP group than that in RIH and normal groups (59.2% vs. 8.7% vs. 9.6%, p < 0.001). Multivariate logistic regression analysis showed that age, diabetes, and smoking history (all p < 0.05) were independent risk factors for RAP. Conclusions In terms of insights gained from OCT, RA atherosclerosis is not uncommon in ACS patients by OCT, sharing several morphological characters with early coronary atherosclerosis. Aging, diabetes, and smoking are risk factors for RAP. Supplementary Information The online version contains supplementary material available at 10.1186/s12872-022-02561-5.
Collapse
Affiliation(s)
- Zixuan Li
- Department of Cardiology, Beijing Luhe Hospital, Capital Medical University, Beijing, China
| | - Zhe Tang
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Yujie Wang
- Department of Cardiology, Beijing Luhe Hospital, Capital Medical University, Beijing, China
| | - Zijing Liu
- Department of Cardiology, Beijing Luhe Hospital, Capital Medical University, Beijing, China
| | - Guozhong Wang
- Department of Cardiology, Beijing Luhe Hospital, Capital Medical University, Beijing, China
| | - Libin Zhang
- Department of Cardiology, Beijing Luhe Hospital, Capital Medical University, Beijing, China
| | - Yongxia Wu
- Department of Cardiology, Beijing Luhe Hospital, Capital Medical University, Beijing, China
| | - Jincheng Guo
- Department of Cardiology, Beijing Luhe Hospital, Capital Medical University, Beijing, China.
| |
Collapse
|
599
|
Cammisotto V, Baratta F, Simeone PG, Barale C, Lupia E, Galardo G, Santilli F, Russo I, Pignatelli P. Proprotein Convertase Subtilisin Kexin Type 9 (PCSK9) Beyond Lipids: The Role in Oxidative Stress and Thrombosis. Antioxidants (Basel) 2022; 11:antiox11030569. [PMID: 35326219 PMCID: PMC8945358 DOI: 10.3390/antiox11030569] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Revised: 03/11/2022] [Accepted: 03/15/2022] [Indexed: 11/16/2022] Open
Abstract
Proprotein convertase subtilisin/kexin type 9 (PCSK9), mainly secreted in the liver, is a key regulator of cholesterol homeostasis inducing LDL receptors’ degradation. Beyond lipid metabolism, PCSK9 is involved in the development of atherosclerosis, promoting plaque formation in mice and human, impairing the integrity of endothelial monolayer and promoting the events that induce atherosclerosis disease progression. In addition, the PCSK9 ancillary role in the atherothrombosis process is widely debated. Indeed, recent evidence showed a regulatory effect of PCSK9 on redox system and platelet activation. In particular, the role of PCSK9 in the activation of nicotinamide adenine dinucleotide phosphate (NADPH) oxidase (Nox2) system, of MAP-kinase cascades and of CD36 and LOX-1 downstream pathways, suggests that PCSK9 may be a significant cofactor in atherothrombosis development. This evidence suggests that the serum levels of PCSK9 could represent a new biomarker for the occurrence of cardiovascular events. Finally, other evidence showed that PCSK9 inhibitors, a novel pharmacological tool introduced in clinical practice in recent years, counteracted these phenomena. In this review, we summarize the evidence concerning the role of PCSK9 in promoting oxidative-stress-related atherothrombotic process.
Collapse
Affiliation(s)
- Vittoria Cammisotto
- Department of Clinical Internal, Anesthesiological and Cardiovascular Sciences, Sapienza University of Rome, 00161 Rome, Italy; (V.C.); (F.B.)
- Department of General Surgery and Surgical Speciality Paride Stefanini, Sapienza University of Rome, 00161 Rome, Italy;
| | - Francesco Baratta
- Department of Clinical Internal, Anesthesiological and Cardiovascular Sciences, Sapienza University of Rome, 00161 Rome, Italy; (V.C.); (F.B.)
| | - Paola G. Simeone
- Department of Medicine and Aging, and Center for Advanced Studies and Technology (CAST), “G. D’Annunzio” University Foundation, 66100 Chieti, Italy; (P.G.S.); (F.S.)
| | - Cristina Barale
- Department of Clinical and Biological Sciences, University of Turin, 10043 Turin, Italy; (C.B.); (I.R.)
| | - Enrico Lupia
- Department of Medical Sciences, University of Turin, 10126 Turin, Italy;
| | - Gioacchino Galardo
- Department of General Surgery and Surgical Speciality Paride Stefanini, Sapienza University of Rome, 00161 Rome, Italy;
| | - Francesca Santilli
- Department of Medicine and Aging, and Center for Advanced Studies and Technology (CAST), “G. D’Annunzio” University Foundation, 66100 Chieti, Italy; (P.G.S.); (F.S.)
| | - Isabella Russo
- Department of Clinical and Biological Sciences, University of Turin, 10043 Turin, Italy; (C.B.); (I.R.)
| | - Pasquale Pignatelli
- Department of Clinical Internal, Anesthesiological and Cardiovascular Sciences, Sapienza University of Rome, 00161 Rome, Italy; (V.C.); (F.B.)
- Mediterranea Cardiocentro, 80133 Napoli, Italy
- Correspondence:
| |
Collapse
|
600
|
Chapman MJ, Zamorano JL, Parhofer KG. Reducing residual cardiovascular risk in Europe: Therapeutic implications of European medicines agency approval of icosapent ethyl/eicosapentaenoic acid. Pharmacol Ther 2022; 237:108172. [PMID: 35304222 DOI: 10.1016/j.pharmthera.2022.108172] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 02/19/2022] [Accepted: 03/10/2022] [Indexed: 12/18/2022]
Abstract
Atherosclerotic cardiovascular disease (ASCVD) and its atherothrombotic complications impose a substantial disease burden in Europe, representing a cost of €210 billion per year for the European Union. Hypertriglyceridemia, a major risk factor for premature ASCVD, is present in more than 20% of the European population, and is a key feature of atherogenic dyslipidemia. Recent findings from the Progression of Early Subclinical Atherosclerosis (PESA) cohort in Spain showed that even in apparently healthy, middle-aged individuals without a history of cardiovascular (CV) risk, elevated triglyceride levels are associated with subclinical atherosclerosis and arterial inflammation. Emerging evidence from epidemiologic and genetic studies supports an independent causative role of triglycerides, triglyceride-rich lipoproteins, and their remnants in this pathology. Icosapent ethyl (IPE) is a highly purified, stable ethyl ester of eicosapentaenoic acid (EPA) that was initially approved by the United States Food and Drug Administration to treat severe hypertriglyceridemia, and subsequently received an expanded indication to reduce the risk of CV events in adult statin-treated patients. Approval was based on the pivotal, randomized, placebo-controlled, double-blind Reduction of Cardiovascular Events with Icosapent Ethyl-Intervention Trial (REDUCE-IT), which showed that high-dose IPE (4 g/day) significantly reduced the risk of primary and secondary composite endpoints comprising major CV events and CV death relative to placebo. In 2021, the European Medicines Agency (EMA) approved IPE to reduce the risk of CV events in adult statin-treated patients at high CV risk with elevated triglyceride levels (≥1.7 mmol/L [≥150 mg/dL]) and established CV disease, or diabetes and at least one other CV risk factor. Clinical studies in Europe, which included patients with acute myocardial infarction, coronary artery disease, and those undergoing cardiac rehabilitation, established that 12.5% to 23.3% of these high-risk populations may benefit from treatment with IPE. Such clinical benefit may in part result from the moderate triglyceride-lowering properties of IPE/EPA; equally however, concentrations of atherogenic remnant particle-cholesterol are markedly reduced. Furthermore, IPE/EPA exerts pleiotropic actions beyond its lipid-lowering properties, which include modulation of endothelial function, attenuation of intra-plaque inflammation and oxidative stress, and reduction in macrophage accumulation. Plasma phospholipids, into which EPA is primarily incorporated and transported, appear to serve as precursors for a series of anti-inflammatory metabolites involving the resolvins RvE1 to RvE3, a pathway which may confer cardioprotective benefits. In addition, plaque imaging data from the Effect of Icosapent Ethyl on Progression of Coronary Atherosclerosis in Patients With Elevated Triglycerides on Statin Therapy (EVAPORATE) and the Combination Therapy of Eicosapentaenoic Acid and Pitavastatin for Coronary Plaque Regression Evaluated by Integrated Backscatter Intravascular Ultrasonography (CHERRY) trials show that plaque stabilization may be favorably affected. These factors may act synergistically to stabilize atherosclerotic plaques and reduce CV risk. In addition to robust efficacy data, multiple cost-utility studies across several countries indicate that IPE/EPA is a cost-effective treatment option that is favorably situated relative to some common willingness-to-pay thresholds. This review will evaluate the relevance of hypertriglyceridemia to residual ASCVD burden in statin-treated dyslipidemic patients, the potential of IPE/EPA to reduce the risk of ASCVD and cardiovascular mortality in high-risk patient populations, and the mechanisms which may underlie these effects. Finally, the clinical implications of the EMA label for IPE will be critically appraised in light of the updated 2019 European Society of Cardiology/European Atherosclerosis Society guidelines on the management of dyslipidemia and the recent European Atherosclerosis Society consensus statement on triglyceride-rich lipoproteins and their remnants, together with considerations of its cost-effectiveness across several countries.
Collapse
Affiliation(s)
- M John Chapman
- Sorbonne University, Endocrinology and Cardiovascular Disease Prevention, Pitié-Salpétrière University Hospital, and National Institute for Health and Medical Research (INSERM), Paris, France.
| | | | - Klaus G Parhofer
- Medical Clinic IV - Grosshadern Hospital of the University of Munich, Munich, Germany
| |
Collapse
|