1
|
Atale N, Wells A. Statins as Secondary Preventive Agent to Limit Breast Cancer Metastatic Outgrowth. Int J Mol Sci 2025; 26:1300. [PMID: 39941069 PMCID: PMC11818786 DOI: 10.3390/ijms26031300] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2024] [Revised: 01/27/2025] [Accepted: 01/31/2025] [Indexed: 02/16/2025] Open
Abstract
Metastasis is a leading cause of mortality in breast cancer, as metastatic disease is often aggressive and resistant to conventional treatments. Cancer cells that spread to distant organs can enter a dormant phase for extended periods, sometimes years or decades. During this dormant phase, cancer cells avoid immune and pharmacological response. Thus, new approaches are needed to prevent these disseminated cells from becoming lethal cancers. Statins are known inhibitors of 3-hydroxy-3-methylglutaryl coenzyme A (HMG-CoA) reductase that have been extensively used in patients with cardiovascular diseases to lower cholesterol. However, recent research has demonstrated their potential in anticancer therapies. Epidemiological evidence suggests that statins are associated with a reduction in breast cancer-specific mortality, although they do not appear to affect the incidence of primary tumors. In this review, we discuss the role of statins in metastasis and dormancy, their cytocidal and cytostatic effects and their interactions with different cell types in the tumor microenvironment. The exact mechanisms by which statins reduce mortality without influencing primary tumor growth remain unclear, also warranting further investigation into their potential role in metastasis and tumor dormancy, which could ultimately help patients to improve survival and quality of life.
Collapse
Affiliation(s)
- Neha Atale
- Department of Pathology, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15213, USA;
| | - Alan Wells
- Department of Pathology, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15213, USA;
- Research and Development Service, Pittsburgh VA Health System, Pittsburgh, PA 15213, USA
- Cell Biology Program, Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA 15213, USA
| |
Collapse
|
2
|
Gholamalizadeh H, Ensan B, Karav S, Jamialahmadi T, Sahebkar A. Regulatory effects of statins on CCL2/CCR2 axis in cardiovascular diseases: new insight into pleiotropic effects of statins. J Inflamm (Lond) 2024; 21:51. [PMID: 39696507 DOI: 10.1186/s12950-024-00420-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: 10/29/2023] [Accepted: 11/13/2024] [Indexed: 12/20/2024] Open
Abstract
BACKGROUND HMG-CoA reductase inhibitors are well-known medications in the treatment of cardiovascular disorders due to their pleiotropic and lipid-lowering properties. Herein, we reviewed the effects of statins on the CCL2/CCR2 axis. METHOD Scopus and Pubmed databases were systematically searched using the following keywords:" Hydroxymethylglutaryl CoA Reductase Inhibitors"," HMG-CoA Reductase Inhibitors"," Statins", "CCL2, Chemokine", "Monocyte Chemoattractant Protein-1" and "Chemokine (C-C Motif) Ligand 2". Evidence investigating the role of statin on MCP-1 in CVD was identified and bibliographies were completely evaluated to gather further related studies. RESULTS The anti-inflammatory effects of statins on the CCL2/CCR2 pathway have been widely investigated. Despite inconclusive results, a great body of research supports the regulatory roles of statins on this pathway due to their pleiotropic effects. By disrupting the CCL2/CCR2 axis, statins attenuate the infiltration of monocytes and macrophages into the zone of inflammation and hence down-regulate the inflammatory cascades in various CVDs including atherosclerosis, cardiac remodeling, and stroke, among others. CONCLUSION CCL2 plays a major role in the pathogenesis of cardiovascular disorders. Down-regulation of CCL2 is proposed as one of the pleiotropic properties of statins. However, more investigations are required to elucidate which statin in what dose exerts a more potent effect on CCL2/CCR2 pathway.
Collapse
Affiliation(s)
- Hanieh Gholamalizadeh
- Student Research Committee, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Behzad Ensan
- Student Research Committee, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Sercan Karav
- Department of Molecular Biology and Genetics, Canakkale Onsekiz Mart University, Canakkale, 17100, Turkey
| | - Tannaz Jamialahmadi
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran.
- Medical Toxicology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
| | - Amirhossein Sahebkar
- Center for Global Health Research, Saveetha Medical College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India.
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran.
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
3
|
Gao X, Luo W, Qu L, Yang M, Chen S, Lei L, Yan S, Liang H, Zhang X, Xiao M, Liao Y, Lee APW, Zhou Z, Chen J, Zhang Q, Wang Y, Xiu J. Genetic association of lipid-lowering drugs with aortic aneurysms: a Mendelian randomization study. Eur J Prev Cardiol 2024; 31:1132-1140. [PMID: 38302118 DOI: 10.1093/eurjpc/zwae044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Revised: 01/28/2024] [Accepted: 01/30/2024] [Indexed: 02/03/2024]
Abstract
AIMS The lack of effective pharmacotherapies for aortic aneurysms (AA) is a persistent clinical challenge. Lipid metabolism plays an essential role in AA. However, the impact of lipid-lowering drugs on AA remains controversial. The study aimed to investigate the genetic association between lipid-lowering drugs and AA. METHODS AND RESULTS Our research used publicly available data on genome-wide association studies (GWASs) and expression quantitative trait loci (eQTL) studies. Genetic instruments, specifically eQTLs related to drug-target genes and SNPs (single nucleotide polymorphisms) located near or within the drug-target loci associated with low-density lipoprotein cholesterol (LDL-C), have been served as proxies for lipid-lowering medications. Drug-Target Mendelian Randomization (MR) study is used to determine the causal association between lipid-lowering drugs and different types of AA. The MR analysis revealed that higher expression of HMGCR (3-hydroxy-3-methylglutaryl coenzyme A reductase) was associated with increased risk of AA (OR = 1.58, 95% CI = 1.20-2.09, P = 1.20 × 10-03) and larger lumen size (aortic maximum area: OR = 1.28, 95% CI = 1.13-1.46, P = 1.48 × 10-04; aortic minimum area: OR = 1.26, 95% CI = 1.21-1.42, P = 1.78 × 10-04). PCSK9 (proprotein convertase subtilisin/kexin type 9) and CETP (cholesteryl ester transfer protein) show a suggestive relationship with AA (PCSK9: OR = 1.34, 95% CI = 1.10-1.63, P = 3.07 × 10-03; CETP: OR = 1.38, 95% CI = 1.06-1.80, P = 1.47 × 10-02). No evidence to support genetically mediated NPC1L1 (Niemann-Pick C1-Like 1) and LDLR (low-density lipoprotein cholesterol receptor) are associated with AA. CONCLUSION This study provides causal evidence for the genetic association between lipid-lowering drugs and AA. Higher gene expression of HMGCR, PCSK9, and CETP increases AA risk. Furthermore, HMGCR inhibitors may link with smaller aortic lumen size.
Collapse
Affiliation(s)
- Xiong Gao
- Department of Cardiovascular Medicine, Nanfang Hospital, Southern Medical University, 1838 Guangzhou Avenue North, Baiyun District, Guangzhou City, Guangdong Province 510515, China
| | - Wei Luo
- Department of Cardiovascular Medicine, Nanfang Hospital, Southern Medical University, 1838 Guangzhou Avenue North, Baiyun District, Guangzhou City, Guangdong Province 510515, China
| | - Liyuan Qu
- Department of Endocrinology, Boluo County People's Hospital, No. 1 Kangbo West Road, Luoyang Street, Boluo County, Huizhou City, Guangdong Province, China
| | - Miaomiao Yang
- Department of Cardiovascular Medicine, Nanfang Hospital, Southern Medical University, 1838 Guangzhou Avenue North, Baiyun District, Guangzhou City, Guangdong Province 510515, China
| | - Siyu Chen
- Department of Cardiovascular Medicine, Nanfang Hospital, Southern Medical University, 1838 Guangzhou Avenue North, Baiyun District, Guangzhou City, Guangdong Province 510515, China
| | - Li Lei
- Department of Cardiology, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), 1017 Dongmen North Road, Luohu District, Shenzhen City, Guangdong Province, China
| | - Shaohua Yan
- Department of Cardiovascular Medicine, Nanfang Hospital, Southern Medical University, 1838 Guangzhou Avenue North, Baiyun District, Guangzhou City, Guangdong Province 510515, China
| | - Hongbin Liang
- Department of Cardiovascular Medicine, Nanfang Hospital, Southern Medical University, 1838 Guangzhou Avenue North, Baiyun District, Guangzhou City, Guangdong Province 510515, China
| | - Xinlu Zhang
- Department of Cardiovascular Medicine, Nanfang Hospital, Southern Medical University, 1838 Guangzhou Avenue North, Baiyun District, Guangzhou City, Guangdong Province 510515, China
| | - Min Xiao
- Department of Cardiovascular Medicine, Nanfang Hospital, Southern Medical University, 1838 Guangzhou Avenue North, Baiyun District, Guangzhou City, Guangdong Province 510515, China
| | - Yulin Liao
- Department of Cardiovascular Medicine, Nanfang Hospital, Southern Medical University, 1838 Guangzhou Avenue North, Baiyun District, Guangzhou City, Guangdong Province 510515, China
| | - Alex Pui-Wai Lee
- Division of Cardiology, Department of Medicine and Therapeutics, Prince of Wales Hospital and Laboratory of Cardiac Imaging and 3D Printing, Li Ka Shing Institute of Health Science, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong Special Administrative Region, China
| | - Zhongjiang Zhou
- Department of Cardiovascular Medicine, Nanfang Hospital, Southern Medical University, 1838 Guangzhou Avenue North, Baiyun District, Guangzhou City, Guangdong Province 510515, China
| | - Jiejian Chen
- Department of Medical Oncology, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, No. 1 Panfu Road, Yuexiu District, Guangzhou City, Guangdong Province, China
| | - Qiuxia Zhang
- Department of Cardiovascular Medicine, Nanfang Hospital, Southern Medical University, 1838 Guangzhou Avenue North, Baiyun District, Guangzhou City, Guangdong Province 510515, China
| | - Yuegang Wang
- Department of Cardiovascular Medicine, Nanfang Hospital, Southern Medical University, 1838 Guangzhou Avenue North, Baiyun District, Guangzhou City, Guangdong Province 510515, China
| | - Jiancheng Xiu
- Department of Cardiovascular Medicine, Nanfang Hospital, Southern Medical University, 1838 Guangzhou Avenue North, Baiyun District, Guangzhou City, Guangdong Province 510515, China
| |
Collapse
|
4
|
Espinosa EVP, Matute EM, Sosa Guzmán DM, Khasawneh FT. The Polypill: A New Alternative in the Prevention and Treatment of Cardiovascular Disease. J Clin Med 2024; 13:3179. [PMID: 38892892 PMCID: PMC11172978 DOI: 10.3390/jcm13113179] [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: 03/18/2024] [Revised: 04/26/2024] [Accepted: 04/27/2024] [Indexed: 06/21/2024] Open
Abstract
Cardiovascular disease (CVD) is the primary cause of death and disability worldwide. Although age-standardized CVD mortality rates decreased globally by 14.5% between 2006 and 2016, the burden of CVD remains disproportionately higher in low- and middle-income countries compared to high-income countries. Even though proven, effective approaches based on multiple-drug intake aimed at the prevention and treatment of CVD are currently available, poor adherence, early discontinuation of treatment, and suboptimal daily execution of the prescribed therapeutic regimes give rise to shortfalls in drug exposure, leading to high variability in the responses to the prescribed medications. Wald and Law, in their landmark paper published in BMJ 2003, hypothesized that the use of a fixed-dose combination of statins, β-blockers, angiotensin receptor blockers, angiotensin-converting enzyme inhibitors, and aspirin (classic Polypill composition) may increase adherence and decrease CVD by up to 80% when prescribed as primary prevention or in substitution of traditional protocols. Since then, many clinical trials have tested this hypothesis, with comparable results. This review aims to describe the available clinical trials performed to assess the impact of fixed-dose combinations on adherence, cost-effectiveness, and the risk factors critical to the onset of CVD.
Collapse
Affiliation(s)
- Enma V. Páez Espinosa
- Department of Clinical Laboratory, School of Medicine, Pontifical Catholic University of Ecuador, Quito 170143, Ecuador;
- Center for Research on Health in Latin America (CISeAL), Pontifical Catholic University of Ecuador, Quito 170143, Ecuador
| | - Eugenia Mato Matute
- Department of Endocrinology and Nutrition, Hospital de la Santa Creu i Sant Pau, 08041 Barcelona, Spain;
- Networking Research Centre of Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), C/Monforte de Lemos 3-5, 28029 Madrid, Spain
| | - Delia M. Sosa Guzmán
- Department of Clinical Laboratory, School of Medicine, Pontifical Catholic University of Ecuador, Quito 170143, Ecuador;
| | - Fadi T. Khasawneh
- Department of Pharmaceutical Sciences, Rangel School of Pharmacy, Texas A&M University, College Station, TX 77843, USA;
| |
Collapse
|
5
|
Kazemi Asl S, Rahimzadegan M, Kazemi Asl A. Pharmacogenomics-based systematic review of coronary artery disease based on personalized medicine procedure. Heliyon 2024; 10:e28983. [PMID: 38601677 PMCID: PMC11004819 DOI: 10.1016/j.heliyon.2024.e28983] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2023] [Revised: 03/27/2024] [Accepted: 03/27/2024] [Indexed: 04/12/2024] Open
Abstract
Background Coronary artery disease (CAD) is the most common reason for mortality and disability-adjusted life years (DALYs) lost globally. This study aimed to suggest a new gene list for the treatment of CAD by a systematic review of bioinformatics analyses of pharmacogenomics impacts of potential genes and variants. Methods PubMed search was filtered by the title including Coronary Artery Disease during 2020-2023. To find the genes with pharmacogenetic impact on the CAD, additional filtrations were considered according to the variant annotations. Protein-Protein Interactions (PPIs), Gene-miRNA Interactions (GMIs), Protein-Drug Interactions (PDIs), and variant annotation assessments (VAAs) performed by STRING-MODEL (ver. 12), Cytoscape (ver. 3.10), miRTargetLink.2., NetworkAnalyst (ver 0.3.0), and PharmGKB. Results Results revealed 5618 publications, 1290 papers were qualified, and finally, 650 papers were included. 4608 protein-coding genes were extracted, among them, 1432 unique genes were distinguished and 530 evidence-based repeated genes remained. 71 genes showed a pharmacogenetics-related variant annotation in at least (entirely 6331 annotations). Variant annotation assessment (VAA) showed 532 potential variants for the final report, and finally, the concluding PGs list represented 175 variants. Based on the function and MAF, 57 nonsynonymous variants of 29 Pharmacogenomics-related genes were associated with CAD. Conclusion Conclusively, evaluating circulating miR33a in individuals' plasma with CAD, and genotyping of rs2230806, rs2230808, rs2487032, rs12003906, rs2472507, rs2515629, and rs4149297 (ABCA1 variants) lead to precisely prescribing of well-known drugs. Also, the findings of this review can be used in both whole-genome sequencing (WGS) and whole-exome sequencing (WES) analysis in the prognosis and diagnosis of CAD.
Collapse
Affiliation(s)
- Siamak Kazemi Asl
- Deputy of Education, Ministry of Health and Medical Education, Tehran, Iran
| | - Milad Rahimzadegan
- Functional Neurosurgery Research Center, Shohada Tajrish Comprehensive Neurosurgical Center of Excellence, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Alireza Kazemi Asl
- Functional Neurosurgery Research Center, Shohada Tajrish Comprehensive Neurosurgical Center of Excellence, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
6
|
Wang HL, Narisawa M, Wu P, Meng X, Cheng XW. The many roles of cathepsins in restenosis. Heliyon 2024; 10:e24720. [PMID: 38333869 PMCID: PMC10850908 DOI: 10.1016/j.heliyon.2024.e24720] [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: 09/01/2023] [Revised: 01/12/2024] [Accepted: 01/12/2024] [Indexed: 02/10/2024] Open
Abstract
Drug-eluting stents (DES) and dual antiplatelet regimens have significantly improved the clinical management of ischemic heart disease; however, the drugs loaded with DES in clinical practice are mostly paclitaxel or rapamycin derivatives, which target symptoms of post implantation proliferation and inflammation, leading to delayed re-endothelialization and neo-atherosclerosis. Along with the treatments already in place, there is a need for novel strategies to lessen the negative clinical outcomes of DES delays as well as a need for greater understanding of their pathobiological mechanisms. This review concentrates on the function of cathepsins (Cats) in the inflammatory response and granulation tissue formation that follow Cat-induced damage to the vasculature scaffold, as well as the functions of Cats in intimal hyperplasia, which is characterized by the migration and proliferation of smooth muscle cells, and endothelial denudation, re-endothelialization, and/or neo-endothelialization. Additionally, Cats can alter essential neointima formation and immune response inside scaffolds, and if Cats are properly controlled in vivo, they may improve scaffold biocompatibility. This unique profile of functions could lead to an original concept for a cathepsin-based coronary intervention treatment as an adjunct to stent placement.
Collapse
Affiliation(s)
- Hai Long Wang
- Department of Adult Intensive Care Unit, Maternal and Child Health Hospital of Hubei Province, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, PR China
- Department of Cardiology and Hypertension, Jilin Provincial Key Laboratory of Stress and Cardiovascular Disease, Yanbian University Hospital, Yanji, Jilin, PR China
| | - Megumi Narisawa
- Department of Cardiology, Nagoya University Graduate School of Medicine, Nagoya, Aichiken, 4668550, Japan
| | - Pan Wu
- Department of Adult Intensive Care Unit, Maternal and Child Health Hospital of Hubei Province, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, PR China
| | - Xiangkun Meng
- Department of Vascular Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310000, PR China
| | - Xian Wu Cheng
- Department of Cardiology and Hypertension, Jilin Provincial Key Laboratory of Stress and Cardiovascular Disease, Yanbian University Hospital, Yanji, Jilin, PR China
- Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, Yanbian University, Yanji, Jilin, 133002, PR China
| |
Collapse
|
7
|
Nemtsova V, Vischer AS, Burkard T. Hypertensive Heart Disease: A Narrative Review Series-Part 3: Vasculature, Biomarkers and the Matrix of Hypertensive Heart Disease. J Clin Med 2024; 13:505. [PMID: 38256639 PMCID: PMC10816030 DOI: 10.3390/jcm13020505] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Revised: 01/11/2024] [Accepted: 01/12/2024] [Indexed: 01/24/2024] Open
Abstract
Over the last few decades, research efforts have resulted in major advances in our understanding of the pathophysiology of hypertensive heart disease (HHD). This is the third part of a three-part review series. Here, we focus on the influence of high blood pressure on the micro- and macroalterations that occur in the vasculature in HHD. We also provide an overview of circulating cardiac biomarkers that may prove useful for a better understanding of the pathophysiology, development and progression of HHD, and may play a unique role in the diagnostic and prognostic evaluation of patients with HHD, taking into account their properties showing as abnormal long before the onset of the disease. In the conclusion, we propose an updated definition of HHD and a matrix for clinical classification, which we suspect will be useful in practice, allowing an individual approach to HHD patients.
Collapse
Affiliation(s)
- Valeriya Nemtsova
- Medical Outpatient Department and Hypertension Clinic, ESH Hypertension Centre of Excellence, University Hospital Basel, 4031 Basel, Switzerland; (V.N.); (A.S.V.)
- Internal Diseases and Family Medicine Department, Educational and Scientific Medical Institute of National Technical University «Kharkiv Polytechnic Institute», 61000 Kharkiv, Ukraine
| | - Annina S. Vischer
- Medical Outpatient Department and Hypertension Clinic, ESH Hypertension Centre of Excellence, University Hospital Basel, 4031 Basel, Switzerland; (V.N.); (A.S.V.)
- Faculty of Medicine, University of Basel, 4056 Basel, Switzerland
| | - Thilo Burkard
- Medical Outpatient Department and Hypertension Clinic, ESH Hypertension Centre of Excellence, University Hospital Basel, 4031 Basel, Switzerland; (V.N.); (A.S.V.)
- Faculty of Medicine, University of Basel, 4056 Basel, Switzerland
- Department of Cardiology, University Hospital Basel, 4031 Basel, Switzerland
| |
Collapse
|
8
|
Hutton M, Frazer M, Lin A, Patel S, Misra A. New Targets in Atherosclerosis: Vascular Smooth Muscle Cell Plasticity and Macrophage Polarity. Clin Ther 2023; 45:1047-1054. [PMID: 37709601 DOI: 10.1016/j.clinthera.2023.08.015] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Revised: 08/07/2023] [Accepted: 08/21/2023] [Indexed: 09/16/2023]
Abstract
PURPOSE Despite an increase in treatment options, and substantial reductions in cardiovascular mortality over the past half-century, atherosclerosis remains the most prevalent cause of premature mortality worldwide. The development of innovative new therapies is crucial to further minimize atherosclerosis-related deaths. The diverse array of cell phenotypes derived from vascular smooth muscle cells (SMCs) and macrophages within atherosclerotic plaques are increasingly becoming recognized for their beneficial and detrimental roles in plaque stability and disease burden. This review explores how contemporary transcriptomics and fate-mapping studies have revealed vascular cell plasticity as a relatively unexplored target for therapeutic intervention. METHODS Recent literature for this narrative review was obtained by searching electronic databases (ie, Google Scholar, PubMed). Additional studies were sourced from reference lists and the authors' personal databases. FINDINGS The lipid-rich and inflammatory plaque milieu induces SMC phenotypic switching to both beneficial and detrimental phenotypes. Likewise, macrophage heterogeneity increases with disease burden to a variety of pro-inflammatory and anti-inflammatory activation states. These vascular cell phenotypes are determinants of plaque structure stability, and it is therefore highly likely that they influence clinical outcomes. Development of clinical treatments targeting deleterious phenotypes or promoting pro-healing phenotypes remains in its infancy. However, existing treatments (statins) have shown beneficial effects toward macrophage polarization, providing a rationale for more targeted approaches. In contrast, beneficial SMC phenotypic modulation with these pharmacologic agents has yet to be achieved. The range of modulated vascular cell phenotypes provides a multitude of novel targets and the potential to reduce future adverse events. IMPLICATIONS Vascular cell phenotypic heterogeneity must continue to be explored to lower cardiovascular events in the future. The rapidly increasing weight of evidence surrounding the role of SMC plasticity and macrophage polarity in plaque vulnerability provides a strong foundation upon which development of new therapeutics must follow. This approach may prove to be crucial in reducing cardiovascular events and improving patient benefit in the future.
Collapse
Affiliation(s)
- Michael Hutton
- Atherosclerosis and Vascular Remodeling Group, Heart Research Institute, Sydney, New South Wales, Australia
| | - Madeleine Frazer
- Atherosclerosis and Vascular Remodeling Group, Heart Research Institute, Sydney, New South Wales, Australia
| | - Alexander Lin
- Atherosclerosis and Vascular Remodeling Group, Heart Research Institute, Sydney, New South Wales, Australia; School of Biomedical Engineering, Faculty of Engineering, The University of Sydney, Sydney, New South Wales, Australia
| | - Sanjay Patel
- Heart Research Institute, The University of Sydney, Sydney, New South Wales, Australia; Royal Prince Alfred Hospital, Sydney, New South Wales, Australia; Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
| | - Ashish Misra
- Atherosclerosis and Vascular Remodeling Group, Heart Research Institute, Sydney, New South Wales, Australia; Heart Research Institute, The University of Sydney, Sydney, New South Wales, Australia; Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia.
| |
Collapse
|
9
|
Puertas-Umbert L, Almendra-Pegueros R, Jiménez-Altayó F, Sirvent M, Galán M, Martínez-González J, Rodríguez C. Novel pharmacological approaches in abdominal aortic aneurysm. Clin Sci (Lond) 2023; 137:1167-1194. [PMID: 37559446 PMCID: PMC10415166 DOI: 10.1042/cs20220795] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Revised: 07/05/2023] [Accepted: 07/28/2023] [Indexed: 08/11/2023]
Abstract
Abdominal aortic aneurysm (AAA) is a severe vascular disease and a major public health issue with an unmet medical need for therapy. This disease is featured by a progressive dilation of the abdominal aorta, boosted by atherosclerosis, ageing, and smoking as major risk factors. Aneurysm growth increases the risk of aortic rupture, a life-threatening emergency with high mortality rates. Despite the increasing progress in our knowledge about the etiopathology of AAA, an effective pharmacological treatment against this disorder remains elusive and surgical repair is still the unique available therapeutic approach for high-risk patients. Meanwhile, there is no medical alternative for patients with small aneurysms but close surveillance. Clinical trials assessing the efficacy of antihypertensive agents, statins, doxycycline, or anti-platelet drugs, among others, failed to demonstrate a clear benefit limiting AAA growth, while data from ongoing clinical trials addressing the benefit of metformin on aneurysm progression are eagerly awaited. Recent preclinical studies have postulated new therapeutic targets and pharmacological strategies paving the way for the implementation of future clinical studies exploring these novel therapeutic strategies. This review summarises some of the most relevant clinical and preclinical studies in search of new therapeutic approaches for AAA.
Collapse
Affiliation(s)
- Lídia Puertas-Umbert
- Institut d’Investigació Biomèdica Sant Pau (IIB SANT PAU), Barcelona, Spain
- CIBER de Enfermedades Cardiovasculares, ISCIII, Madrid, Spain
| | | | - Francesc Jiménez-Altayó
- Department of Pharmacology, Therapeutics and Toxicology, School of Medicine, Universitat Autònoma de Barcelona, Barcelona, Spain
- Neuroscience Institute, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Marc Sirvent
- CIBER de Enfermedades Cardiovasculares, ISCIII, Madrid, Spain
- Departamento de Angiología y Cirugía Vascular del Hospital Universitari General de Granollers, Granollers, Barcelona, Spain
| | - María Galán
- Institut d’Investigació Biomèdica Sant Pau (IIB SANT PAU), Barcelona, Spain
- CIBER de Enfermedades Cardiovasculares, ISCIII, Madrid, Spain
- Departamento de Ciencias Básicas de la Salud, Universidad Rey Juan Carlos, Alcorcón, Spain
| | - José Martínez-González
- Institut d’Investigació Biomèdica Sant Pau (IIB SANT PAU), Barcelona, Spain
- CIBER de Enfermedades Cardiovasculares, ISCIII, Madrid, Spain
- Instituto de Investigaciones Biomédicas de Barcelona (IIBB-CSIC), Barcelona, Spain
| | - Cristina Rodríguez
- Institut d’Investigació Biomèdica Sant Pau (IIB SANT PAU), Barcelona, Spain
- CIBER de Enfermedades Cardiovasculares, ISCIII, Madrid, Spain
| |
Collapse
|
10
|
Mangion D, Pace NP, Formosa MM. The relationship between adipokine levels and bone mass-A systematic review. Endocrinol Diabetes Metab 2023; 6:e408. [PMID: 36759562 PMCID: PMC10164433 DOI: 10.1002/edm2.408] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Revised: 01/10/2023] [Accepted: 01/14/2023] [Indexed: 02/11/2023] Open
Abstract
INTRODUCTION Adipose tissue is the source of a broad array of signalling molecules (adipokines), which mediate interorgan communication and regulate metabolic homeostasis. Alterations in adipokine levels have been causally implicated in various metabolic disorders, including changes in bone mass. Osteoporosis is the commonest progressive metabolic bone disease, characterized by elevated risk of fragility fractures as a result of a reduced bone mass and microarchitectural deterioration. The effects of different adipokines on bone mass have been studied in an attempt to identify novel modulators of bone mass or diagnostic biomarkers of osteoporosis. METHODS In this review, we sought to aggregate and assess evidence from independent studies that quantify specific adipokines and their effect on bone mineral density (BMD). RESULTS A literature search identified 57 articles that explored associations between different adipokines and BMD. Adiponectin and leptin were the most frequently studied adipokines, with most studies demonstrating that adiponectin levels are associated with decreased BMD at the lumbar spine and femoral neck. Conversely, leptin levels are associated with increased BMD at these sites. However, extensive heterogeneity with regards to sample size, characteristics of study subjects, ethnicity, as well as direction and magnitude of effect at specific skeletal anatomical sites was identified. The broad degree of conflicting findings reported in this study can be attributed several factors. These include differences in study design and ascertainment criteria, the analytic challenges of quantifying specific adipokines and their isoforms, pre-analytical variables (in particular patient preparation) and confounding effects of co-existing disease. CONCLUSIONS This review highlights the biological relevance of adipokines in bone metabolism and reinforces the need for longitudinal research to elucidate the causal relationship of adipokines on bone mass.
Collapse
Affiliation(s)
- Darren Mangion
- Department of Applied Biomedical Science, Faculty of Health Sciences, University of Malta, Msida, Malta
| | - Nikolai P Pace
- Centre for Molecular Medicine and Biobanking, University of Malta, Msida, Malta.,Department of Anatomy, Faculty of Medicine and Surgery, University of Malta, Msida, Malta
| | - Melissa M Formosa
- Department of Applied Biomedical Science, Faculty of Health Sciences, University of Malta, Msida, Malta.,Centre for Molecular Medicine and Biobanking, University of Malta, Msida, Malta
| |
Collapse
|
11
|
Li R, Liu Y, Jiang J. Research advances in drug therapy for abdominal aortic aneurysms over the past five years: An updated narrative review. Int J Cardiol 2023; 372:93-100. [PMID: 36462700 DOI: 10.1016/j.ijcard.2022.11.058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/27/2022] [Revised: 11/16/2022] [Accepted: 11/27/2022] [Indexed: 12/03/2022]
Abstract
BACKGROUND Abdominal aortic aneurysms (AAA) rupture can lead to patient death. Surgical treatment is currently the optimal treatment for AAA with large diameter (≥50 mm). For AAA with small diameter (30-50 mm), how to administer effective pharmacological treatment to reduce aneurysm expansion rate and rupture risk is the current focus in the field of vascular surgery. There is still no effective drug for the treatment of asymptomatic AAA. METHODS This article searches the PubMed, Web of Science, Embase, and Cochrane databases for clinical studies on the drug treatment of abdominal aortic aneurysms in the past 5 years. The latest progress in the drug treatment of AAA was reviewed, including antibiotics, antihypertensive drugs, antiplatelet drugs, hypoglycemic drugs, hypolipidemic drugs, mast cell inhibitors and corticosteroids. RESULTS 25 studies were included in this narrative review. Among them, metformin revealed therapeutic effect in 2 prospective cohort study and 3 retrospective cohort study. The therapeutic effect of statins was controversial in 3 retrospective cohort study. However, the definite therapeutic effects of antihypertensive agents, antibiotics, mast cell inhibitors, antiplatelet agents and corticosteroids on abdominal aortic aneurysms have not been verified in prospective studies. CONCLUSION Metformin provided a positive effect in reducing expansion rate, rupture risk, and perioperative mortality. The therapeutic effect of statins was controversial, which warrant further validation in prospective cohorts. However, there is still a lack of effective agents for the treatment of AAA based on recent studies.
Collapse
Affiliation(s)
- Ruihua Li
- Department of General Surgery, Vascular Surgery, Qilu Hospital of Shandong University, No.107, Road Wen Hua Xi, Jinan, Shandong 250012, China.
| | - Yang Liu
- Department of General Surgery, Vascular Surgery, Qilu Hospital of Shandong University, No.107, Road Wen Hua Xi, Jinan, Shandong 250012, China.
| | - Jianjun Jiang
- Department of General Surgery, Vascular Surgery, Qilu Hospital of Shandong University, No.107, Road Wen Hua Xi, Jinan, Shandong 250012, China.
| |
Collapse
|
12
|
Yu SY, Li HL, Tse YK, Li X, Ren QW, Wu MZ, Wong PF, Tse HF, Lip GYH, Yiu KH. Pre-admission and In-Hospital Statin Use is Associated With Reduced Short-Term Mortality in Infective Endocarditis. Mayo Clin Proc 2023; 98:252-265. [PMID: 36114025 DOI: 10.1016/j.mayocp.2022.06.006] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/27/2021] [Revised: 04/11/2022] [Accepted: 06/02/2022] [Indexed: 02/07/2023]
Abstract
OBJECTIVE To investigate for potential protective effects of statin use among patients with infective endocarditis (IE) with consideration for underlying diseases and bacterial culture - variables which have prognostic implications and show considerable geographic variation yet are unappreciated in previous pharmacoepidemiological studies. PATIENTS AND METHODS Patients diagnosed with IE between January 1, 1996, and December 31, 2019, were identified. We estimated the effect on mortality of pre-admission statin use (≥90 cumulative days of use before index date) and in-hospital use (use beginning within 2 days of admission), compared with nonusers and discontinued users, respectively, through propensity score analytics. RESULTS Of 6700 IE patients (mean age, 58.0 years; 63.3% male [n=4251]), 776 patients had pre-admission statin use, with 626 continuing statin use following admission (in-hospital users). Pre-admission statin users had a 31% lower risk of 1-year mortality (HR, 0.69; 95% CI, 0.58 to 0.82) compared with nonusers. In-hospital users had a 48% lower risk of 1-year mortality (HR, 0.52; 95% CI, 0.34 to 0.78) compared with discontinued users. Subgroup analyses showed significant protective effects of statin use for patients with varying causative agents, underlying diseases, and with or without prosthetic valves. Results were consistent across different statins, and were dose-dependent. CONCLUSION In patients with IE, pre-admission and in-hospital use of statin, when compared with statin nonusers and discontinued users, respectively, were associated with a lower risk of 1-year mortality.
Collapse
Affiliation(s)
- Si-Yeung Yu
- Division of Cardiology, Department of Medicine, the University of Hong Kong Shenzhen Hospital, Shenzhen, China; Division of Cardiology, Department of Medicine, the University of Hong Kong, Queen Mary Hospital, Hong Kong, China
| | - Hang-Long Li
- Division of Cardiology, Department of Medicine, the University of Hong Kong, Queen Mary Hospital, Hong Kong, China
| | - Yi-Kei Tse
- Division of Cardiology, Department of Medicine, the University of Hong Kong, Queen Mary Hospital, Hong Kong, China
| | - Xue Li
- Division of Cardiology, Department of Medicine, the University of Hong Kong, Queen Mary Hospital, Hong Kong, China
| | - Qing-Wen Ren
- Division of Cardiology, Department of Medicine, the University of Hong Kong Shenzhen Hospital, Shenzhen, China; Division of Cardiology, Department of Medicine, the University of Hong Kong, Queen Mary Hospital, Hong Kong, China
| | - Mei-Zhen Wu
- Division of Cardiology, Department of Medicine, the University of Hong Kong Shenzhen Hospital, Shenzhen, China; Division of Cardiology, Department of Medicine, the University of Hong Kong, Queen Mary Hospital, Hong Kong, China
| | - Pui-Fai Wong
- Division of Cardiology, Department of Medicine, the University of Hong Kong, Queen Mary Hospital, Hong Kong, China
| | - Hung-Fat Tse
- Division of Cardiology, Department of Medicine, the University of Hong Kong Shenzhen Hospital, Shenzhen, China; Division of Cardiology, Department of Medicine, the University of Hong Kong, Queen Mary Hospital, Hong Kong, China
| | - Gregory Y H Lip
- Liverpool Centre for Cardiovascular Science, University of Liverpool and Liverpool Heart & Chest Hospital, Liverpool, United Kingdom
| | - Kai-Hang Yiu
- Division of Cardiology, Department of Medicine, the University of Hong Kong Shenzhen Hospital, Shenzhen, China; Division of Cardiology, Department of Medicine, the University of Hong Kong, Queen Mary Hospital, Hong Kong, China.
| |
Collapse
|
13
|
Urbanowicz T, Olasińska-Wiśniewska A, Michalak M, Perek B, Al-Imam A, Rodzki M, Witkowska A, Straburzyńska-Migaj E, Bociański M, Misterski M, Lesiak M, Jemielity M. Pre-operative systemic inflammatory response index influences long-term survival rate in off-pump surgical revascularization. PLoS One 2022; 17:e0276138. [PMID: 36520919 PMCID: PMC9754600 DOI: 10.1371/journal.pone.0276138] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Accepted: 09/29/2022] [Indexed: 12/23/2022] Open
Abstract
Coronary artery bypass revascularization is still the optimal treatment for complex coronary artery disease with good long-term results. The relation between inflammatory activation in the post-operative period and the long-term prognosis was already postulated. The possible predictive role of preoperative inflammatory indexes after the off-pump coronary artery bypass grafting technique on long term survival was the aim of the study. Study population included 171 patients with a median age of 64 years (59-64) operated on using off-pump technique between January and December 2014. Patients enrolled in the current study were followed-up for 8 years. We conducted a multivariable analysis of pre-operative and post-operative inflammatory markers based on analysis of the whole blood count. The overall survival rate was 80% for the total follow-up period, while 34 deaths were reported (30-days mortality rate of 1%). In the multivariable analysis, a pre-operative value of systemic inflammatory response index (SIRI) >1.27 (HR = 6.16, 95% CI 2.17-17.48, p = 0.012) revealed a prognostic value for long-term mortality assessment after off-pump surgery. Preoperative inflammatory activation evaluated by systemic inflammatory reaction index (SIRI) possess a prognostic value for patients with complex coronary artery disease. The SIRI value above 1.27 indicates a worse late prognosis after off-pump coronary artery bypass (AUC = 0.682, p<0.001).
Collapse
Affiliation(s)
- Tomasz Urbanowicz
- Cardiac Surgery and Transplantology Department, Poznan University of Medical Sciences, Poznan, Poland
- * E-mail:
| | - Anna Olasińska-Wiśniewska
- Cardiac Surgery and Transplantology Department, Poznan University of Medical Sciences, Poznan, Poland
| | - Michał Michalak
- Department of Computer Science and Statistics, Poznan University of Medical Sciences, Poznan, Poland
| | - Bartłomiej Perek
- Cardiac Surgery and Transplantology Department, Poznan University of Medical Sciences, Poznan, Poland
| | - Ahmed Al-Imam
- Department of Computer Science and Statistics, Poznan University of Medical Sciences, Poznan, Poland
- Department of Anatomy and Cellular Biology, College of Medicine, University of Baghdad, Baghdad, Iraq
| | - Michał Rodzki
- Cardiac Surgery and Transplantology Department, Poznan University of Medical Sciences, Poznan, Poland
| | - Anna Witkowska
- Cardiac Surgery and Transplantology Department, Poznan University of Medical Sciences, Poznan, Poland
| | | | - Michał Bociański
- Cardiac Surgery and Transplantology Department, Poznan University of Medical Sciences, Poznan, Poland
| | - Marcin Misterski
- Cardiac Surgery and Transplantology Department, Poznan University of Medical Sciences, Poznan, Poland
| | - Maciej Lesiak
- 1 Cardiology Department, Poznan University of Medical Sciences, Poznan, Poland
| | - Marek Jemielity
- Cardiac Surgery and Transplantology Department, Poznan University of Medical Sciences, Poznan, Poland
| |
Collapse
|
14
|
Hosseini A, Sahranavard T, Reiner Ž, Jamialahmadi T, Dhaheri YA, Eid AH, Sahebkar A. Effect of statins on abdominal aortic aneurysm. Eur J Pharm Sci 2022; 178:106284. [PMID: 36038100 DOI: 10.1016/j.ejps.2022.106284] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Revised: 07/21/2022] [Accepted: 08/24/2022] [Indexed: 11/30/2022]
Abstract
Abdominal aortic aneurysm (AAA) is a prevalent condition which causes progressive growth and rupture of aortic wall with a high death rate. Several studies have found that treatment with statins may decrease the progress of AAA and the risk of rupture by suppressing the inflammatory mediators, decreasing oxidative stress, and inhibiting mechanisms involved in extracellular matrix (ECM) degradation. Moreover, some studies have reported that prehospital therapy with statins can decrease mortality after surgery. The novelty of this paper is that different studies including those performed in humans and animals were reviewed and the potential mechanisms by which statins can have an effect on AAA were summarized. Overall, the evidence suggested an association between treatment with statins and improvement of AAA.
Collapse
Affiliation(s)
- Azar Hosseini
- Pharmacological Research Center of Medicinal Plants, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Toktam Sahranavard
- Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Željko Reiner
- Department of Internal Medicine, University Hospital Center Zagreb, School of Medicine University of Zagreb, Zagreb, Croatia
| | - Tannaz Jamialahmadi
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Yusra Al Dhaheri
- Department of Biology, College of Science, United Arab Emirates University, AlAin, United Arab Emirates
| | - Ali H Eid
- Department of Basic Medical Sciences, College of Medicine, QU Health, Qatar University, Doha, Qatar.
| | - Amirhossein Sahebkar
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran; Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Biotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
15
|
Que Y, Zhang Z, Zhang Y, Li X, Chen L, Chen P, Ou C, Yang C, Chang J. Silicate ions as soluble form of bioactive ceramics alleviate aortic aneurysm and dissection. Bioact Mater 2022; 25:716-731. [PMID: 37056259 PMCID: PMC10086764 DOI: 10.1016/j.bioactmat.2022.07.005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Revised: 06/21/2022] [Accepted: 07/05/2022] [Indexed: 12/15/2022] Open
Abstract
Aortic aneurysm and dissection (AAD) are leading causes of death in the elderly. Recent studies have demonstrated that silicate ions can manipulate multiple cells, especially vascular-related cells. We demonstrated in this study that silicate ions as soluble form of bioactive ceramics effectively alleviated aortic aneurysm and dissection in both Ang II and β-BAPN induced AAD models. Different from the single targeting therapeutic drug approaches, the bioactive ceramic derived approach attributes to the effect of bioactive silicate ions on the inhibition of the AAD progression through regulating the local vascular microenvironment of aorta systematically in a multi-functional way. The in vitro experiments revealed that silicate ions did not only alleviate senescence and inflammation of the mouse aortic endothelial cells, enhance M2 polarization of mouse bone marrow-derived macrophages, and reduce apoptosis of mouse aortic smooth muscle cells, but also regulate their interactions. The in vivo studies further confirm that silicate ions could effectively alleviate senescence, inflammation, and cell apoptosis of aortas, accomplished with reduced aortic dilation, collagen deposition, and elastin laminae degradation. This bioactive ceramic derived therapy provides a potential new treatment strategy in attenuating AAD progression.
Collapse
|
16
|
Su Z, Guo J, Gu Y. Pharmacotherapy in Clinical Trials for Abdominal Aortic Aneurysms: A Systematic Review and Meta-Analysis. Clin Appl Thromb Hemost 2022; 28:10760296221120423. [PMID: 36083182 PMCID: PMC9465599 DOI: 10.1177/10760296221120423] [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] [Indexed: 11/15/2022] Open
Abstract
OBJECTIVE There is no medical treatment proven to limit abdominal aortic aneurysm (AAA) progression. This systematic review aimed to summarise available trial evidence on the efficacy of pharmacotherapy in limiting AAA growth and AAA-related events. METHODS A systematic literature search was performed to examine the efficacy of pharmacotherapy in reducing AAA growth and AAA-related events. Pubmed, Embase (Excerpta Medica Database), and the Cochrane library were searched from March, 1999 to March 29, 2022. AAA growth (mm/year) in the intervention and control groups was expressed as mean and standard deviation (SD). The results of AAA growth were expressed as mean difference (MD) and its 95% confidence interval (95% CI). Odds ratios (ORs) were calculated for the AAA-related events.Heterogeneity was quantified using the I2 statistic. Forest plots were created to show the pooled results of each outcome. OUTCOMES A total of 1373 articles were found in different databases according to the search strategy, and 10 articles were identified by hand searching. A total of 26 articles were included in our systematic review after the screening. For the studies of metformin, the meta-analysis demonstrated that metformin use was associated with a lower AAA growth rate (MD: -0.81 mm/y, 95% CI: -1.19 to -0.42, P < 0.0001, I2 = 87%), Metformin use also was related to the lower rates of AAA-related events (OR: 0.53, 95% CI: 0.36 to 0.76, P = 0.0007, I2 = 60%). The hypotensive drugs of the studies mainly included angiotensin-converting enzyme inhibitors (ACEI), angiotensin II type 1 receptor blockers (ARB), and propranolol. The overall meta-analysis of blood pressure-lowering drugs reported no significant effect in limiting the AAA growth (MD: 0.31mm/year, 95%CI: -0.03 to 0.65, P = 0.07, I2 = 66%) and AAA-related events (OR: 1.33, 95%CI: 0.76 to 2.32, P = 0.32, I2 = 98%), In the subgroup analysis of the hypotensive drugs, the ACEI/ARB and propranolol also showed no significant in reducing the AAA growth and AAA-related events. The meta-analysis of the antibiotics demonstrated that the antibiotics were not associated with a lower AAA growth rate (MD: -0.27 mm/y, 95% CI: -0.88 to 0.34, P = 0.39, I2 = 77%) and AAA-related events (OR: 0.94, 95%CI: 0.65 to 1.35, P = 0.72, I2 = 0%). The results of statins also showed no significant effect in limiting AAA growth (MD: -1.11mm/year, 95%CI: -2.38 to 0.16, P = 0.09, I2 = 96%) and AAA-related events (OR: 0.53, 95%CI: 0.26 to 1.06, P = 0.07, I2 = 92%). CONCLUSION In conclusion, effective pharmacotherapy for AAA was still lacking. Although the meta-analysis showed that metformin use was associated with lower AAA growth and AAA-related events, all of the included studies about metformin were cohort studies or case-control studies. More randomized controlled trials (RCTs) are needed for further verification.
Collapse
Affiliation(s)
- Zhixiang Su
- Vascular Surgery Department, 71044Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Jianming Guo
- Vascular Surgery Department, 71044Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Yongquan Gu
- Vascular Surgery Department, 71044Xuanwu Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
17
|
Lee JS, Park SC, Kim SD. Effects of hypercholesterolism on expansion of abdominal aortic aneurysm in rat model. J Cardiothorac Surg 2021; 16:352. [PMID: 34961565 PMCID: PMC8711175 DOI: 10.1186/s13019-021-01734-1] [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: 07/08/2021] [Accepted: 11/24/2021] [Indexed: 11/25/2022] Open
Abstract
Background Inflammation is recognized as a critical process in expansion of abdominal aortic aneurysm (AAA). A relationship between effects of cholesterol and statin in this process have been suggested, but remain untested. Therefore, current study aimed to examine the effects of hypercholesterolism on expansion of AAA in a rat model. Methods A total of 16 male rats were divided into 4 groups as follows: group I, normocholesterol diet and saline infusion, group II, normocholesterol diet and porcine pancreatic elastase (PPE) infusion, group III, hypercholesterol diet and PPE infusion, and group IV, hypercholesterol diet, PPE infusion and statin administration. At the 3rd week, saline was infused intraluminally in group I and PPE in groups II-IV to induce AAA. At the 5th week, blood and aortic tissue were obtained from each rat for evaluation of lipid profiles, aortic diameters (ADs), and characteristics of stains. Results Post-procedural aortic diameter (AD3) and AD3/pre-procedural aortic diameter (AD1) were significantly different among four groups (P = 0.042, P = 0.028, respectively). AD3 was significantly larger in group II than group I, and group III than group IV (P = 0.012, P = 0.043, respectively). AD3/AD1 was significantly higher in group II than group I, and group III than group II (P = 0.008, P = 0.030, respectively). Group III showed the highest cellularity for inflammatory cells. Conclusions Though larger experimental and clinical studies are necessary, authors suggest that hypercholesterolism can aggravate expansion of AAA, and that statin therapy can reduce it. Therefore, monitoring for hypercholesterolism and instituting statin therapy may be helpful to suppress expansion of AAA.
Collapse
Affiliation(s)
- Jong Seok Lee
- Division of Vascular and Transplant Surgery, Department of Surgery, College of Medicine, The Catholic University of Korea, Seoul, 06591, Korea
| | - Sun Cheol Park
- Division of Vascular and Transplant Surgery, Department of Surgery, College of Medicine, The Catholic University of Korea, Seoul, 06591, Korea
| | - Sang Dong Kim
- Division of Vascular and Transplant Surgery, Department of Surgery, College of Medicine, The Catholic University of Korea, Seoul, 06591, Korea. .,Division of Vascular and Transplant Surgery, Department of Surgery, Incheon St. Mary's Hospital, The Catholic University of Korea, 56 Dongsu-ro, Bupyong-Gu, Incheon, 21431, Korea.
| |
Collapse
|
18
|
Statins: Neurobiological underpinnings and mechanisms in mood disorders. Neurosci Biobehav Rev 2021; 128:693-708. [PMID: 34265321 DOI: 10.1016/j.neubiorev.2021.07.012] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Revised: 06/28/2021] [Accepted: 07/10/2021] [Indexed: 12/26/2022]
Abstract
Statins (3-hydroxy-3-methylglutaryl-coenzyme A reductase inhibitors) treat dyslipidaemia and cardiovascular disease by inhibiting cholesterol biosynthesis. They also have immunomodulatory and anti-inflammatory properties. Beyond cardiovascular disease, cholesterol and inflammation appear to be components of the pathogenesis and pathophysiology of neuropsychiatric disorders. Statins may therefore afford some therapeutic benefit in mood disorders. In this paper, we review the pathophysiology of mood disorders with a focus on pharmacologically relevant pathways, using major depressive disorder and bipolar disorder as exemplars. Statins are discussed in the context of these disorders, with particular focus on the putative mechanisms involved in their anti-inflammatory and immunomodulatory effects. Recent clinical data suggest that statins may have antidepressant properties, however given their interactions with many known biological pathways, it has not been fully elucidated which of these are the major determinants of clinical outcomes in mood disorders. Moreover, it remains unclear what the appropriate dose, or appropriate patient phenotype for adjunctive treatment may be. High quality randomised control trials in concert with complementary biological investigations are needed if the potential clinical effects of statins on mood disorders, as well as their biological correlates, are to be better understood.
Collapse
|
19
|
Statins reduce mortality after abdominal aortic aneurysm repair: A systematic review and meta-analysis. J Vasc Surg 2021; 75:356-362.e4. [PMID: 34197945 DOI: 10.1016/j.jvs.2021.06.033] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Accepted: 06/01/2021] [Indexed: 12/17/2022]
Abstract
OBJECTIVE The necessity and efficacy of statin treatment for abdominal aortic aneurysm (AAA) remains controversial. This systematic review and meta-analysis was conducted to investigate the effects of statin therapy on the outcomes of patients with AAA. METHODS The Cochrane library, Embase, and MedLine were searched comprehensively to identify relevant cohort studies and randomized controlled trials. The primary outcomes included short- and long-term mortality after AAA repair, and secondary outcomes included the incidence of perioperative cardiovascular complications, sac shrinkage after endovascular aneurysm repair, and the growth rate of the aneurysms. Short-term mortality was defined as all-cause 30-day or in-hospital postoperative mortality. Long-term mortality was defined as the all-cause mortality at the end of follow-up period (≥1 year). A random effects model was used to combine the results of included studies. Forest plots were created to show the pooled results of each outcome. RESULTS One post hoc analysis of a randomized trial and 36 cohort studies (n = 134,290 patients) were included in this systematic review. The average score of included studies by Newcastle-Ottawa Scale was 7.76. Patients taking or not taking statin therapy were all diagnosed with unruptured AAA, and 59.9% of these patients were given statin therapy. Compared with statin nonusers, patients in statin therapy had significantly lower long-term mortality (odds ratio, 0.67; 95% confidence interval, 0.59-0.75; P < .001; I2 = 71.7%), and short-term mortality after aneurysmal repair (odds ratio, 0.51; 95% confidence interval, 0.36-0.73; P < .001; I2 = 81.4%). No significant difference was found between patients taking or not taking statin treatment on perioperative cardiovascular complications or sac shrinkage after endovascular aneurysm repair or growth rate of AAA under surveillance. CONCLUSIONS These findings suggest that statin use is associated with a significant decrease in long- and short-term mortality in patients after AAA repair. Based on these results, statin therapy is worth being used in clinical practice for the management of AAA.
Collapse
|
20
|
Weininger G, Chan SM, Zafar M, Ziganshin BA, Elefteriades JA. Risk reduction and pharmacological strategies to prevent progression of aortic aneurysms. Expert Rev Cardiovasc Ther 2021; 19:619-631. [PMID: 34102944 DOI: 10.1080/14779072.2021.1940958] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
INTRODUCTION While size thresholds exist to determine when aortic aneurysms warrant surgical intervention, there is no consensus on how best to treat this disease before aneurysms reach the threshold for intervention. Since a landmark study in 1994 first suggested ß-blockers may be useful in preventing aortic aneurysm growth, there has been a surge in research investigating different pharmacologic therapies for aortic aneurysms - with very mixed results. AREAS COVERED We have reviewed the existing literature on medical therapies used for thoracic and abdominal aortic aneurysms in humans. These include ß-blockers, angiotensin II receptor blockers, and angiotensin-converting enzyme inhibitors as well as miscellaneous drugs such as tetracyclines, macrolides, statins, and anti-platelet medications. EXPERT OPINION While multiple classes of drugs have been explored for risk reduction in aneurysm disease, with few exceptions results have been disappointing with an abundance of contradictory findings. The vast majority of studies have been done in patients with abdominal aortic aneurysms or thoracic aortic aneurysm patients with Marfan Syndrome. There exists a striking gap in the literature when it comes to pharmacologic management of non-Marfan Syndrome patients with thoracic aortic aneurysms. Given the differences in pathogenesis, this is an important future direction for aortic aneurysm research.
Collapse
Affiliation(s)
- Gabe Weininger
- Aortic Institute at Yale-New Haven Hospital, Yale University School of Medicine, New Haven, CT, USA
| | - Shin Mei Chan
- Aortic Institute at Yale-New Haven Hospital, Yale University School of Medicine, New Haven, CT, USA
| | - Mohammad Zafar
- Aortic Institute at Yale-New Haven Hospital, Yale University School of Medicine, New Haven, CT, USA
| | - Bulat A Ziganshin
- Aortic Institute at Yale-New Haven Hospital, Yale University School of Medicine, New Haven, CT, USA
| | - John A Elefteriades
- Aortic Institute at Yale-New Haven Hospital, Yale University School of Medicine, New Haven, CT, USA
| |
Collapse
|
21
|
Meekel JP, Dias-Neto M, Bogunovic N, Conceição G, Sousa-Mendes C, Stoll GR, Leite-Moreira A, Huynh J, Micha D, Eringa EC, Balm R, Blankensteijn JD, Yeung KK. Inflammatory Gene Expression of Human Perivascular Adipose Tissue in Abdominal Aortic Aneurysms. Eur J Vasc Endovasc Surg 2021; 61:1008-1016. [PMID: 33858751 DOI: 10.1016/j.ejvs.2021.02.034] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2020] [Revised: 02/15/2021] [Accepted: 02/20/2021] [Indexed: 01/08/2023]
Abstract
OBJECTIVE Perivascular adipose tissue (PVAT) contributes to vascular homeostasis and is increasingly linked to vascular pathology. PVAT density and volume were associated with abdominal aortic aneurysm (AAA) presence and dimensions on imaging. However, mechanisms underlying the role of PVAT in AAA have not been clarified. This study aimed to explore differences in PVAT from AAA using gene expression and functional tests. METHODS Human aortic PVAT and control subcutaneous adipose tissue were collected during open AAA surgery. Gene analyses and functional tests were performed. The control group consisted of healthy aorta from non-living renal transplant donors. Gene expression tests were performed to study genes potentially involved in various inflammatory processes and AAA related genes. Live PVAT and subcutaneous adipose tissue (SAT) from AAA were used for ex vivo co-culture with smooth muscle cells (SMCs) retrieved from non-pathological aortas. RESULTS Adipose tissue was harvested from 27 AAA patients (n [gene expression] = 22, n [functional tests] = 5) and five control patients. An increased inflammatory gene expression of PTPRC (p = .008), CXCL8 (p = .033), LCK (p = .003), CCL5 (p = .004) and an increase in extracellular matrix breakdown marker MMP9 (p = .016) were found in AAA compared with controls. Also, there was a decreased anti-inflammatory gene expression of PPARG in AAA compared with controls (p = .040). SMC co-cultures from non-pathological aortas with PVAT from AAA showed increased MMP9 (p = .033) and SMTN (p = .008) expression and SAT increased SMTN expression in these SMC. CONCLUSION The data revealed that PVAT from AAA shows an increased pro-inflammatory and matrix metallopeptidase gene expression and decreased anti-inflammatory gene expression. Furthermore, increased expression of genes involved in aneurysm formation was found in healthy SMC co-culture with PVAT of AAA patients. Therefore, PVAT from AAA might contribute to inflammation of the adjacent aortic wall and thereby plays a possible role in AAA pathophysiology. These proposed pathways of inflammatory induction could reveal new therapeutic targets in AAA treatment.
Collapse
Affiliation(s)
- Jorn P Meekel
- Department of Vascular Surgery, Amsterdam University Medical Centres, location VUmc, Amsterdam, the Netherlands; Department of Physiology, Amsterdam University Medical Centres, Amsterdam Cardiovascular Sciences, Amsterdam, the Netherlands; Department of Surgery, Zaans Medisch Centrum, Zaandam, the Netherlands
| | - Marina Dias-Neto
- Department of Angiology and Vascular Surgery, São João University Hospital Centre, Porto, Portugal; Department of Surgery and Physiology, Cardiovascular Research Unit, Faculty of Medicine, University of Porto, Portugal
| | - Natalija Bogunovic
- Department of Vascular Surgery, Amsterdam University Medical Centres, location VUmc, Amsterdam, the Netherlands; Department of Physiology, Amsterdam University Medical Centres, Amsterdam Cardiovascular Sciences, Amsterdam, the Netherlands
| | - Gloria Conceição
- Department of Surgery and Physiology, Cardiovascular Research Unit, Faculty of Medicine, University of Porto, Portugal
| | - Claudia Sousa-Mendes
- Department of Surgery and Physiology, Cardiovascular Research Unit, Faculty of Medicine, University of Porto, Portugal
| | - Gawin R Stoll
- Department of Physiology, Amsterdam University Medical Centres, Amsterdam Cardiovascular Sciences, Amsterdam, the Netherlands
| | - Adelino Leite-Moreira
- Department of Surgery and Physiology, Cardiovascular Research Unit, Faculty of Medicine, University of Porto, Portugal
| | - Jennifer Huynh
- Department of Vascular Surgery, Amsterdam University Medical Centres, location VUmc, Amsterdam, the Netherlands; Department of Physiology, Amsterdam University Medical Centres, Amsterdam Cardiovascular Sciences, Amsterdam, the Netherlands
| | - Dimitra Micha
- Department of Clinical Genetics, Amsterdam University Medical Centres, location VUmc, Amsterdam, the Netherlands
| | - Etto C Eringa
- Department of Physiology, Amsterdam University Medical Centres, Amsterdam Cardiovascular Sciences, Amsterdam, the Netherlands
| | - Ron Balm
- Department of Vascular Surgery, Amsterdam University Medical Centres, location AMC, Amsterdam, the Netherlands
| | - Jan D Blankensteijn
- Department of Vascular Surgery, Amsterdam University Medical Centres, location VUmc, Amsterdam, the Netherlands
| | - Kak K Yeung
- Department of Vascular Surgery, Amsterdam University Medical Centres, location VUmc, Amsterdam, the Netherlands; Department of Physiology, Amsterdam University Medical Centres, Amsterdam Cardiovascular Sciences, Amsterdam, the Netherlands; Department of Vascular Surgery, Amsterdam University Medical Centres, location AMC, Amsterdam, the Netherlands.
| |
Collapse
|
22
|
Risum Ø, Sandven I, Sundhagen JO, Abdelnoor M. Editor's Choice – Effect of Statins on Total Mortality in Abdominal Aortic Aneurysm Repair: A Systematic Review and Meta-analysis. Eur J Vasc Endovasc Surg 2021; 61:114-120. [DOI: 10.1016/j.ejvs.2020.08.007] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2019] [Revised: 07/03/2020] [Accepted: 08/05/2020] [Indexed: 01/22/2023]
|
23
|
Statin use is associated with lower disease severity in COVID-19 infection. Sci Rep 2020; 10:17458. [PMID: 33060704 PMCID: PMC7562925 DOI: 10.1038/s41598-020-74492-0] [Citation(s) in RCA: 79] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Accepted: 09/29/2020] [Indexed: 12/30/2022] Open
Abstract
We aim to study the association of hyperlipidemia and statin use with COVID-19 severity. We analysed a retrospective cohort of 717 patients admitted to a tertiary centre in Singapore for COVID-19 infection. Clinical outcomes of interest were oxygen saturation ≤ 94% requiring supplemental oxygen, intensive-care unit (ICU) admission, invasive mechanical-ventilation and death. Patients on long term dyslipidaemia medications (statins, fibrates or ezetimibe) were considered to have dyslipidaemia. Logistic regression models were used to study the association between dyslipidaemia and clinical outcomes adjusted for age, gender and ethnicity. Statin treatment effect was determined, in a nested case-control design, through logistic treatment models with 1:3 propensity matching for age, gender and ethnicity. All statistical tests were two-sided, and statistical significance was taken as p < 0.05. One hundred fifty-six (21.8%) patients had dyslipidaemia and 97% of these were on statins. Logistic treatment models showed a lower chance of ICU admission for statin users when compared to non-statin users (ATET: Coeff (risk difference): - 0.12 (- 0.23, - 0.01); p = 0.028). There were no other significant differences in other outcomes. Statin use was independently associated with lower ICU admission. This supports current practice to continue prescription of statins in COVID-19 patients.
Collapse
|
24
|
Hyun MH, Jang JW, Lee E, An H, Seog Seo H. Baseline, delta, and achieved low-density lipoprotein cholesterol levels and cardiovascular risk in patients on statin therapy: A post-hoc resampling mediation analysis of treating new targets [TNT] trial. Clin Exp Pharmacol Physiol 2020; 47:1649-1658. [PMID: 32583886 DOI: 10.1111/1440-1681.13367] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Revised: 04/01/2020] [Accepted: 06/10/2020] [Indexed: 11/30/2022]
Abstract
Clinical guidelines for monitoring low-density lipoprotein cholesterol (LDL-C) after statin therapy do not clearly define the clinical roles of baseline LDL-C, ΔLDL-C, and achieved LDL-C according to statin intensity. We performed post-hoc analysis of the Treating to New Target (TNT) study to evaluate individual LDL-C parameters after statin therapy. Primary outcome was the risk for total major adverse cardiovascular events (MACE). We use resampling multilevel mediation analysis to analyze complex relationships among LDL-C parameters based on similar statin intensities. Tertiles for resample A (matched baseline LDL-C; distinct achieved LDL), resample B (matched ΔLDL-C; distinct baseline LDL-C), and resample C (matched achieved LDL-C; distinct ΔLDL-C) were analyzed using Cox proportional hazard ratios. In original data analysis, the incidence of MACE was reduced in those with lower achieved LDL-C in total, low, and high intensity statin users (hazard ratios [HRs] = 0.990, 0.992, 0.992; respectively; all P-values < .001). In mediation analysis, resample A showed consistently high incidence for MACE in the middle tertile (HR = 1.237; 95% confidential interval [CI] = 1.008-1.517; P-value = .041) and highest tertile (HR = 1.275; 95% CI = 1.021-1.592; P-value = .032) compared to the lowest tertile. However, resamples B and C did not show consistent differences. Similarly, no consistent statistical difference in MACE according to statin intensity. Lower achieved LDL-C decreased MACE in participants with a similar baseline LDL-C after statin therapy. However, the change in absolute values of ΔLDL-C and achieved LDL-C should be interpreted in an individualized manner due to their complex collinearity, and statin intensity should also be taken into consideration.
Collapse
Affiliation(s)
- Myung Han Hyun
- Department of Internal Medicine, Korea University Medical Center, Seoul, Korea
| | - Jae Won Jang
- Department of Biostatistics, Korea University College of Medicine, Seoul, Korea
| | - Eunmi Lee
- Division of Cardiology, Department of Internal Medicine, Wonkwang University Sanbon Hospital, Gunpo, Korea
| | - Hyonggin An
- Department of Biostatistics, Korea University College of Medicine, Seoul, Korea
| | - Hong Seog Seo
- Division of Cardiology, Department of Internal Medicine, Korea University Guro Hospital, Seoul, Korea
| |
Collapse
|
25
|
Song TJ, Kim J. Effect of Statins on the Risk of Poststroke Pneumonia: National Population-Based Cohort Study. Infect Drug Resist 2020; 13:2689-2698. [PMID: 32982323 PMCID: PMC7493019 DOI: 10.2147/idr.s258420] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Accepted: 07/12/2020] [Indexed: 12/28/2022] Open
Abstract
Purpose Statins are widely prescribed medications for treatment of dyslipidemia and prevention of cardiovascular disease. Beyond their lipid-lowering property, statins exhibit multiple pleiotropic and antimicrobial effects. We aimed to investigate the effect of statins on the long-term risk of pneumonia after acute ischemic stroke. Methods This retrospective observational research was performed using South Korean National Health Insurance Service claim data, which consist of population-based random sampling. We included patients discharged with acute ischemic stroke (I63 in the ICD10) and no prior history of pneumonia. The primary outcome measure was the occurrence of pneumonia determined based on ICD10 code J09–J18. Treatment with statins during follow-up was collected as a time-dependent variable based on prescription records. Results A total of 7,001 subjects with acute ischemic stroke and no prior history of pneumonia were included. During the mean 3.96-year follow-up, pneumonia occurred in 1,715 subjects (24.5%). On multivariate time-dependent Cox proportional hazard–regression analyses, significant preventive benefit of treatment with statins against pneumonia was noted (adjusted HR 0.86, 95% CI 0.77–0.97). Compared to no use of statin, adjusted HRs (95% CIs) for current use of low–intermediate high-intensity statins were 0.88 (0.78–0.99) and 0.49 (0.27–0.87), respectively. Conclusion Our retrospective national cohort study found reduced risk of poststroke pneumonia with statin therapy after acute ischemic stroke. Our study suggests that treatment with statins may have a preventive effect against the common complication of poststroke pneumonia.
Collapse
Affiliation(s)
- Tae-Jin Song
- Department of Neurology, Seoul Hospital, Ewha Womans University College of Medicine, Seoul 07804, South Korea
| | - Jinkwon Kim
- Department of Neurology, Yongin Severance Hospital, Yonsei University College of Medicine, Yongin 16995, South Korea.,Department of Neurology, CHA Bundang Medical Center, CHA University, Seongnam 13496, South Korea
| |
Collapse
|
26
|
Sladojevic M, Zlatanovic P, Stanojevic Z, Koncar I, Vidicevic S, Tasic J, Isakovic A, Tomic I, Mutavdzic P, Stevanovic K, Trailovic R, Davidovic L. Influence of preoperative statins and aspirin administration on biological and magnetic resonance imaging properties in patients with abdominal aortic aneurysm. VASA 2020; 50:116-124. [PMID: 32669062 DOI: 10.1024/0301-1526/a000895] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Background: Main objective of this study was to evaluate the influence of statins and/or acetylsalicylic acid on biochemical characteristics of abdominal aortic aneurysm (AAA) wall and intraluminal thrombus (ILT). Patients and methods: Fifty patients with asymptomatic infrarenal AAA were analyzed using magnetic resonance imaging on T1w sequence. Relative ILT signal intensity (SI) was determined as a ratio between ILT and psoas muscle SI. Samples containing the full ILT thickness and aneurysm wall were harvested from the anterior surface at the level of the maximal diameter. The concentration of enzymes such as matrix metalloproteinase (MMP) 9, MMP2 and neutrophil elastase (NE/ELA) were analyzed in ILT and AAA wall; while collagen type III, elastin and proteoglycan 4 were analyzed in harvested AAA wall. Oxidative stress in the AAA wall was assessed by catalase and malondialdehyde activity in tissue samples. Results: Relative ILT signal intensity (1.09 ± 0.41 vs 0.89 ± 0.21, p = 0.013) were higher in non-statin than in statin group. Patients who were taking aspirin had lower relative ILT area (0.89 ± 0.19 vs 1.13. ± 0.44, p = 0.016), and lower relative ILT signal intensity (0.85 [0.73-1.07] vs 1.01 [0.84-1.19], p = 0.021) compared to non-aspirin group. There were higher concentrations of elastin in AAA wall among patients taking both of aspirin and statins (1.21 [0.77-3.02] vs 0.78 (0.49-1.05) ng/ml, p = 0.044) than in patients who did not take both of these drugs. Conclusions: Relative ILT SI was lower in patients taking statin and aspirin. Combination of antiplatelet therapy and statins was associated with higher elastin concentrations in AAA wall.
Collapse
Affiliation(s)
- Milos Sladojevic
- School of Medicine, University of Belgrade, Serbia.,Clinic for Vascular and Endovascular Surgery, Clinical Center of Serbia, Belgrade, Serbia
| | - Petar Zlatanovic
- Clinic for Vascular and Endovascular Surgery, Clinical Center of Serbia, Belgrade, Serbia
| | - Zeljka Stanojevic
- Institute of Medical and Clinical Biochemistry, School of Medicine, University of Belgrade, Serbia
| | - Igor Koncar
- School of Medicine, University of Belgrade, Serbia.,Clinic for Vascular and Endovascular Surgery, Clinical Center of Serbia, Belgrade, Serbia
| | - Sasenka Vidicevic
- Institute of Medical and Clinical Biochemistry, School of Medicine, University of Belgrade, Serbia
| | - Jelena Tasic
- Institute of Medical and Clinical Biochemistry, School of Medicine, University of Belgrade, Serbia
| | - Aleksandra Isakovic
- Clinic for Vascular and Endovascular Surgery, Clinical Center of Serbia, Belgrade, Serbia.,Institute of Medical and Clinical Biochemistry, School of Medicine, University of Belgrade, Serbia
| | - Ivan Tomic
- School of Medicine, University of Belgrade, Serbia.,Clinic for Vascular and Endovascular Surgery, Clinical Center of Serbia, Belgrade, Serbia
| | - Perica Mutavdzic
- Clinic for Vascular and Endovascular Surgery, Clinical Center of Serbia, Belgrade, Serbia
| | - Ksenija Stevanovic
- School of Medicine, University of Belgrade, Serbia.,Clinic for Vascular and Endovascular Surgery, Clinical Center of Serbia, Belgrade, Serbia
| | - Ranko Trailovic
- Clinic for Vascular and Endovascular Surgery, Clinical Center of Serbia, Belgrade, Serbia
| | - Lazar Davidovic
- School of Medicine, University of Belgrade, Serbia.,Clinic for Vascular and Endovascular Surgery, Clinical Center of Serbia, Belgrade, Serbia
| |
Collapse
|
27
|
Agrawal H, Choy HHK, Liu J, Auyoung M, Albert MA. Coronary Artery Disease. Arterioscler Thromb Vasc Biol 2020; 40:e185-e192. [PMID: 32579480 DOI: 10.1161/atvbaha.120.313608] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Harsh Agrawal
- From the Center for the Study of Adversity and Cardiovascular Disease (NURTURE Center), Division of Cardiology, Department of Medicine, University of California San Francisco (H.A., M.A.A.)
| | - Ho-Hin K Choy
- Division of Cardiology, Department of Medicine, California Pacific Medical Center, San Francisco (H.-h.K.C., J.L., M.A.)
| | - Jason Liu
- Division of Cardiology, Department of Medicine, California Pacific Medical Center, San Francisco (H.-h.K.C., J.L., M.A.)
| | - Matthew Auyoung
- Division of Cardiology, Department of Medicine, California Pacific Medical Center, San Francisco (H.-h.K.C., J.L., M.A.)
| | - Michelle A Albert
- From the Center for the Study of Adversity and Cardiovascular Disease (NURTURE Center), Division of Cardiology, Department of Medicine, University of California San Francisco (H.A., M.A.A.)
| |
Collapse
|
28
|
Liberale L, Carbone F, Montecucco F, Sahebkar A. Statins reduce vascular inflammation in atherogenesis: A review of underlying molecular mechanisms. Int J Biochem Cell Biol 2020; 122:105735. [PMID: 32126319 DOI: 10.1016/j.biocel.2020.105735] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2019] [Revised: 01/23/2020] [Accepted: 02/28/2020] [Indexed: 01/09/2023]
Abstract
Chronic inflammation enhances the detrimental role of dyslipidaemia during atherogenesis. Statins are among the most effective anti-atherosclerotic medications, being able to impact on both cardiovascular morbidity and mortality. Although these molecules have been first described as lipid-lowering medications, several lines of evidence suggest additional benefits through their "pleiotropic" anti-atherosclerotic activities. Specifically, statins can modulate vascular atherosclerotic inflammation by directly improving functions of endothelial cells, vascular smooth muscle cells, platelets, and immune cells. Here, we discuss basic and clinical evidence to provide an update on the molecular mechanisms underlying the protective anti-inflammatory role of statins in atherogenesis.
Collapse
Affiliation(s)
- Luca Liberale
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, 6 viale Benedetto XV, 16132 Genoa, Italy; Center for Molecular Cardiology, University of Zurich, Wagistrasse 12, 8952 Schlieren, Switzerland.
| | - Federico Carbone
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, 6 viale Benedetto XV, 16132 Genoa, Italy; IRCCS Ospedale Policlinico San Martino Genoa - Italian Cardiovascular Network, 10 Largo Benzi, 16132, Genoa, Italy
| | - Fabrizio Montecucco
- IRCCS Ospedale Policlinico San Martino Genoa - Italian Cardiovascular Network, 10 Largo Benzi, 16132, Genoa, Italy; First Clinic of Internal Medicine, Department of Internal Medicine and Centre of Excellence for Biomedical Research (CEBR), University of Genoa, 6 viale Benedetto XV, 16132 Genoa, Italy
| | - Amirhossein Sahebkar
- Halal Research Center of IRI, FDA, Tehran, Iran; Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Neurogenic Inflammation Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
29
|
Impact of BDNF Val66Met Polymorphism on Myocardial Infarction: Exploring the Macrophage Phenotype. Cells 2020; 9:cells9051084. [PMID: 32349267 PMCID: PMC7290372 DOI: 10.3390/cells9051084] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Revised: 04/22/2020] [Accepted: 04/24/2020] [Indexed: 12/13/2022] Open
Abstract
Brain-derived neurotrophic factor (BDNF) is a member of the neurotrophin growth factor family, well known for its role in the homeostasis of the cardiovascular system. Recently, the human BDNF Val66Met single nucleotide polymorphism has been associated with the increased propensity for arterial thrombosis related to acute myocardial infarction (AMI). Using cardiac magnetic resonance imaging and immunohistochemistry analyses, we showed that homozygous mice carrying the human BDNF Val66Met polymorphism (BDNFMet/Met) undergoing left anterior descending (LAD) coronary artery ligation display an adverse cardiac remodeling compared to wild-type (BDNFVal/Val). Interestingly, we observed a persistent presence of pro-inflammatory M1-like macrophages and a reduced accumulation of reparative-like phenotype macrophages (M2-like) in the infarcted heart of mutant mice. Further qPCR analyses showed that BDNFMet/Met peritoneal macrophages are more pro-inflammatory and have a higher migratory ability compared to BDNFVal/Val ones. Finally, macrophages differentiated from circulating monocytes isolated from BDNFMet/Met patients with coronary heart disease displayed the same pro-inflammatory characteristics of the murine ones. In conclusion, the BDNF Val66Met polymorphism predisposes to adverse cardiac remodeling after myocardial infarction in a mouse model and affects macrophage phenotype in both humans and mice. These results provide a new cellular mechanism by which this human BDNF genetic variant could influence cardiovascular disease.
Collapse
|
30
|
Chia PY, Thein TL, Ong SWX, Lye DC, Leo YS. Severe dengue and liver involvement: an overview and review of the literature. Expert Rev Anti Infect Ther 2020; 18:181-189. [PMID: 31971031 DOI: 10.1080/14787210.2020.1720652] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Introduction: Two billion population are at risk of dengue fever and by 2080, over six billion population will be at risk. Hepatitis is common in dengue and the liver is invariably involved in severe cases. We conducted a literature review using the PubMed database on articles covering a broad range of issues related to dengue and hepatitis.Areas covered: This article overviews available literature on changes in the definition of severe dengue, pathogenesis of liver involvement in dengue, clinical manifestations, and predictors of mortality in severe dengue with liver involvement, impact of viral hepatitis co-infections and hepatotoxic drugs, and hemophagocytic lymphohistiocytosis.Expert commentary: Hepatitis is commonly seen in dengue however the degree of elevation of transaminases did not correlate well with severity of illness in observational studies, except in the elderly. The underlying pathogenesis of liver injury is still being elucidated and further studies are required to fully understand the cellular pathways. Acute or chronic viral hepatitis does not appear to affect dengue outcomes. Commonly used medications such as paracetamol and statins may influence dengue outcomes.
Collapse
Affiliation(s)
- Po Ying Chia
- National Centre for Infectious Diseases, Singapore, Singapore.,Department of Infectious Diseases, Tan Tock Seng Hospital, Singapore, Singapore.,Lee Kong Chian School of Medicine, Singapore, Singapore
| | - Tun-Linn Thein
- National Centre for Infectious Diseases, Singapore, Singapore
| | - Sean Wei Xiang Ong
- National Centre for Infectious Diseases, Singapore, Singapore.,Department of Infectious Diseases, Tan Tock Seng Hospital, Singapore, Singapore
| | - David Chien Lye
- National Centre for Infectious Diseases, Singapore, Singapore.,Department of Infectious Diseases, Tan Tock Seng Hospital, Singapore, Singapore.,Lee Kong Chian School of Medicine, Singapore, Singapore.,Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Yee Sin Leo
- National Centre for Infectious Diseases, Singapore, Singapore.,Department of Infectious Diseases, Tan Tock Seng Hospital, Singapore, Singapore.,Lee Kong Chian School of Medicine, Singapore, Singapore.,Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.,Saw Swee Hock School of Public Health, National University of Singapore, Singapore, Singapore
| |
Collapse
|
31
|
The role of IL-1β in aortic aneurysm. Clin Chim Acta 2020; 504:7-14. [PMID: 31945339 DOI: 10.1016/j.cca.2020.01.007] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2019] [Revised: 01/07/2020] [Accepted: 01/08/2020] [Indexed: 12/12/2022]
Abstract
Interleukin-1β (IL-1β) is a vital cytokine that plays an important role in regulating immune responses to infectious challenges and sterile insults. In addition, two endogenous inhibitors of functional receptor binding, IL-1 receptor antagonist (IL-1Ra), complete the family. To gain biological activity, IL-1β requires processing by the protease caspase-1 and activation of inflammasomes. Numerous clinical association studies and experimental approaches have implicated members of the IL-1 family, their receptors, or components of the processing machinery in the underlying processes of cardiovascular diseases. Here, we summarize the current state of knowledge regarding the pro-inflammatory and disease-modulating role of the IL-1 family in aneurysm. We discuss clinical evidence, signalling pathway, and mechanism of action and last, lend a perspective on currently developing therapeutic strategies involving IL-1β in aneurysm.
Collapse
|
32
|
Zanetti HR, Gonçalves A, Teixeira Paranhos Lopes L, Mendes EL, Roever L, Silva-Vergara ML, Neves FF, Resende ES. Effects of Exercise Training and Statin Use in People Living with Human Immunodeficiency Virus with Dyslipidemia. Med Sci Sports Exerc 2020; 52:16-24. [PMID: 31834252 DOI: 10.1249/mss.0000000000002120] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
PURPOSE To evaluate the effects of the combination of exercise training (ET) and statins in people living with human immunodeficiency virus. METHODS This was a randomized, double-blind, placebo-controlled clinical trial. Eighty-three people living with human immunodeficiency virus were assigned to either placebo (PL), statins (STA), PL + ET (PLET) or STA + ET (STAET) groups. Volunteers assigned to STA and STAET groups were administered 10 mg of rosuvastatin, whereas the PL and PLET groups were administered a placebo. The PLET and STAET groups performed ET three times a week. Before and after the 12-wk follow-up, the volunteers underwent to anthropometric assessment and blood collection to evaluate lipid profile, cardiovascular markers, inflammatory profile; a Doppler ultrasound examination, muscle strength (MS) and cardiorespiratory fitness (CF) tests were performed. RESULTS There was a decrease in total cholesterol, triglycerides, low-density lipoprotein, C-reactive protein, fibrinogen, interleukin (IL)-1β and right carotid intima-media thickness in the STA, PLET, and STAET groups compared with PL group (P < 0.001). Furthermore, there was a decrease in total cholesterol, triglycerides, low-density lipoprotein, IL-1β, IL-6, and IL-8 levels and in left and right carotid intima-media thickness and an increase in HDL-c levels in the STAET groups compared with the STA (P ≤ 0.001) and PLET groups (P ≤ 0.001). There was an increase in IL-10 levels, peak-systolic velocity, end-diastolic velocity, wall shear rate in the PLET and STAET groups compared with the PL (P ≤ 0.001) and STA groups (P ≤ 0.001). The PLET and STAET groups reduced body fat mass, body fat percentage and increased lean body mass, MS and CF compared with PL (P ≤ 0.001) and STA (P ≤ 0.001) groups. CONCLUSIONS The combination of ET and statins is useful to enhance lipid and inflammatory profiles, reduce cardiovascular disease markers, and improve Doppler ultrasound findings, MS and CF in people living with HIV.
Collapse
Affiliation(s)
| | | | | | - Edmar Lacerda Mendes
- Institute of Health Sciences, Department of Sport Sciences, Federal University of Triângulo Mineiro, Uberaba/MG, BRAZIL
| | - Leonardo Roever
- Postgraduate Program in Health Science, Faculty of Medicine, Federal University of Uberlândia, Uberlândia/MG, BRAZIL
| | - Mário Leon Silva-Vergara
- Institute of Health Sciences, Department of Medical Clinics, Clinics Hospital, Federal University of Triângulo Mineiro, Uberaba/MG, BRAZIL
| | - Fernando Freitas Neves
- Institute of Health Sciences, Department of Medical Clinics, Clinics Hospital, Federal University of Triângulo Mineiro, Uberaba/MG, BRAZIL
| | - Elmiro Santos Resende
- Postgraduate Program in Health Science, Faculty of Medicine, Federal University of Uberlândia, Uberlândia/MG, BRAZIL
| |
Collapse
|
33
|
Roles of Achieved Levels of Low-Density Lipoprotein Cholesterol and High-Sensitivity C-Reactive Protein on Cardiovascular Outcome in Statin Therapy. Cardiovasc Ther 2019; 2019:3824823. [PMID: 31885691 PMCID: PMC6906885 DOI: 10.1155/2019/3824823] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2019] [Revised: 08/16/2019] [Accepted: 08/29/2019] [Indexed: 12/13/2022] Open
Abstract
In statin therapy, the prognostic role of achieved low-density lipoprotein cholesterol (LDL-C) and high-sensitivity C-reactive protein (hsCRP) in cardiovascular outcomes has not been fully elucidated. A total of 4,803 percutaneous coronary intervention (PCI)-naïve patients who prescribed moderate intensity of statin therapy were followed up. Total and each component of major adverse cardiovascular events (MACE) according to LDL-C and hsCRP quartiles were compared. The incidence of 5-year total MACEs in the highest quartile group according to the followed-up hsCRP was higher than that in the lowest quartile (hazard ratio (HR) = 2.16, p < 0.001). However, there was no difference between the highest and lowest quartiles of the achieved LDL-C (HR = 0.95, p = 0.743). After adjustment of potential confounders, the incidence of total death, de novo PCI, atrial fibrillation, and heart failure in the highest quartile of followed-up hsCRP, was higher than that in the lowest quartile (all p < 0.05). However, other components except for de novo PCI in the highest quartile by achieved LDL-C was not different to that in the lowest quartile. These results suggest that followed-up hsCRP can be more useful for predicting future cardiovascular outcome than achieved LDL-C in PCI-naïve patients with statin therapy.
Collapse
|
34
|
Salata K, Syed M, Hussain MA, de Mestral C, Greco E, Mamdani M, Tu JV, Forbes TL, Bhatt DL, Verma S, Al-Omran M. Statins Reduce Abdominal Aortic Aneurysm Growth, Rupture, and Perioperative Mortality: A Systematic Review and Meta-Analysis. J Am Heart Assoc 2019; 7:e008657. [PMID: 30371297 PMCID: PMC6404894 DOI: 10.1161/jaha.118.008657] [Citation(s) in RCA: 84] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Background There are no recognized pharmacological treatments for abdominal aortic aneurysms (AAA), although statins are suggested to be beneficial. We sought to summarize the literature regarding the effects of statins on human AAA growth, rupture, and 30‐day mortality. Methods and Results We conducted a systematic review and meta‐analysis of randomized and observational studies using the Cochrane CENTRAL database, MEDLINE, and EMBASE up to June 15, 2018. Review, abstraction, and quality assessment were conducted by 2 independent reviewers, and a third author resolved discrepancies. Pooled mean differences and odds ratios with 95% confidence intervals were calculated using random effects models. Heterogeneity was quantified using the I2 statistic, and publication bias was assessed using funnel plots. Our search yielded 911 articles. One case‐control and 21 cohort studies involving 80 428 patients were included. The risk of bias was low to moderate. Statin use was associated with a mean AAA growth rate reduction of 0.82 mm/y (95% confidence interval 0.33, 1.32, P=0.001, I2=86%). Statins were also associated with a lower rupture risk (odds ratio 0.63, 95% confidence interval 0.51, 0.78, P<0.0001, I2=27%), and preoperative statin use was associated with a lower 30‐day mortality following elective AAA repair (odds ratio 0.55, 95% confidence interval 0.36, 0.83, P=0.005, I2=57%). Conclusions Statin therapy may be associated with reduction in AAA progression, rupture, and lower rates of perioperative mortality following elective AAA repair. These data argue for widespread statin use in AAA patients. Clinical Trial Registration URL: http://www.crd.york.ac.uk. Unique identifier: CRD42017056480.
Collapse
Affiliation(s)
- Konrad Salata
- 1 Division of Vascular Surgery Department of Surgery University of Toronto Ontario Canada.,2 Division of Vascular Surgery Li Ka Shing Knowledge Institute of St. Michael's Hospital Toronto Ontario Canada
| | - Muzammil Syed
- 3 Faculty of Science McMaster University Hamilton Ontario Canada
| | - Mohamad A Hussain
- 1 Division of Vascular Surgery Department of Surgery University of Toronto Ontario Canada.,2 Division of Vascular Surgery Li Ka Shing Knowledge Institute of St. Michael's Hospital Toronto Ontario Canada
| | - Charles de Mestral
- 1 Division of Vascular Surgery Department of Surgery University of Toronto Ontario Canada.,2 Division of Vascular Surgery Li Ka Shing Knowledge Institute of St. Michael's Hospital Toronto Ontario Canada
| | - Elisa Greco
- 1 Division of Vascular Surgery Department of Surgery University of Toronto Ontario Canada.,2 Division of Vascular Surgery Li Ka Shing Knowledge Institute of St. Michael's Hospital Toronto Ontario Canada
| | - Muhammad Mamdani
- 4 Li Ka Shing Centre for Healthcare Analytics Research and Training (CHART) Li Ka Shing Knowledge Institute St. Michael's Hospital Toronto Ontario Canada.,5 Leslie Dan Faculty of Pharmacy University of Toronto Ontario Canada.,6 Department of Medicine Faculty of Medicine University of Toronto Ontario Canada.,7 Institute of Health Policy, Management and Evaluation Dalla Lana Faculty of Public Health University of Toronto Ontario Canada.,8 Institute for Clinical Evaluative Sciences at Sunnybrook Hospital Toronto Ontario Canada
| | - Jack V Tu
- 7 Institute of Health Policy, Management and Evaluation Dalla Lana Faculty of Public Health University of Toronto Ontario Canada.,8 Institute for Clinical Evaluative Sciences at Sunnybrook Hospital Toronto Ontario Canada.,9 Division of Cardiology Department of Medicine Schulich Heart Program Sunnybrook Hospital Toronto Ontario Canada.,10 Schulich Heart Research Program Sunnybrook Research Institute at Sunnybrook Hospital Toronto Ontario Canada
| | - Thomas L Forbes
- 1 Division of Vascular Surgery Department of Surgery University of Toronto Ontario Canada.,11 Division of Vascular Surgery Toronto General Hospital Toronto Ontario Canada
| | - Deepak L Bhatt
- 12 Brigham and Women's Hospital Heart and Vascular Center Boston MA.,13 Harvard Medical School Boston MA
| | - Subodh Verma
- 14 Division of Cardiac Surgery Department of Surgery University of Toronto Ontario Canada.,15 Division of Cardiac Surgery Li Ka Shing Knowledge Institute of St. Michael's Hospital Toronto Ontario Canada
| | - Mohammed Al-Omran
- 1 Division of Vascular Surgery Department of Surgery University of Toronto Ontario Canada.,2 Division of Vascular Surgery Li Ka Shing Knowledge Institute of St. Michael's Hospital Toronto Ontario Canada.,16 Department of Surgery King Saud University Riyadh Kingdom of Saudi Arabia
| |
Collapse
|
35
|
Lunde NN, Bosnjak T, Solberg R, Johansen HT. Mammalian legumain – A lysosomal cysteine protease with extracellular functions? Biochimie 2019; 166:77-83. [DOI: 10.1016/j.biochi.2019.06.002] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2019] [Accepted: 06/04/2019] [Indexed: 12/31/2022]
|
36
|
Enhanced endoplasmic reticulum and mitochondrial stress in abdominal aortic aneurysm. Clin Sci (Lond) 2019; 133:1421-1438. [PMID: 31239294 DOI: 10.1042/cs20190399] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2019] [Revised: 06/22/2019] [Accepted: 06/24/2019] [Indexed: 02/08/2023]
Abstract
Abdominal aortic aneurysm (AAA) is a degenerative vascular disease with a complex aetiology that remains to be fully elucidated. Clinical management of AAA is limited to surgical repair, while an effective pharmacotherapy is still awaited. Endoplasmic reticulum (ER) stress and mitochondrial dysfunction have been involved in the pathogenesis of cardiovascular diseases (CVDs), although their contribution to AAA development is uncertain. Therefore, we aimed to determine their implication in AAA and investigated the profile of oxysterols in plasma, specifically 7-ketocholesterol (7-KC), as an ER stress inducer.In the present study, we determined aortic ER stress activation in a large cohort of AAA patients compared with healthy donors. Higher gene expression of activating transcription factor (ATF) 6 (ATF6), IRE-1, X-binding protein 1 (XBP-1), C/EBP-homologous protein (CHOP), CRELD2 and suppressor/enhancer of Lin-12-like (SEL1L) and greater protein levels of active ATF6, active XBP1 and of the pro-apoptotic protein CHOP were detected in human aneurysmatic samples. This was accompanied by an exacerbated apoptosis, higher reactive oxygen species (ROS) production and by a reduction in mitochondrial biogenesis in the vascular wall of AAA. The quantification of oxysterols, performed by liquid chromatography-(atmospheric pressure chemical ionization (APCI))-mass spectrometry, showed that levels of 7-KC were significantly higher while those of 7α-hydroxycholesterol (HC), 24-HC and 27-HC were lower in AAA patients compared with healthy donors. Interestingly, the levels of 7-KC correlate with the expression of ER stress markers.Our results evidence an induction of ER stress in the vascular wall of AAA patients associated with an increase in circulating 7-KC levels and a reduction in mitochondrial biogenesis suggesting their implication in the pathophysiology of this disease.
Collapse
|
37
|
Arterial Hypertension and Interleukins: Potential Therapeutic Target or Future Diagnostic Marker? Int J Hypertens 2019; 2019:3159283. [PMID: 31186952 PMCID: PMC6521461 DOI: 10.1155/2019/3159283] [Citation(s) in RCA: 85] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2019] [Accepted: 03/26/2019] [Indexed: 02/06/2023] Open
Abstract
Hypertension as a multifactorial pathology is one of the most important cardiovascular risk factors, affecting up to 30-40% of the general population. Complex immune responses are involved in the inflammatory mechanism of hypertension, with evidence pointing to increased inflammatory mediators even in prehypertensive patients. Increased vascular permeability, thrombogenesis, and fibrosis, effects that are associated with sustained hypertension, could be attributed to chronic inflammation. Chronic inflammation triggers endothelial dysfunction via increased production of ROS through proinflammatory cytokines. Increased serum level of proinflammatory cytokines such as IL-1β, IL-6, IL-8, IL-17, IL-23, TGFβ, and TNFα in hypertensive patients has been associated with either increased blood pressure values and/or end-organ damage. Moreover, some cytokines (i.e., IL-6) seem to determine a hypertensive response to angiotensin II, regardless of blood pressure values. Understanding hypertension as an inflammatory-based pathology gives way to new therapeutic targets. As such, conventional cardiovascular drugs (statins, calcium channels blockers, and ACEIs/ARBs) have shown additional anti-inflammatory effects that could be linked to their blood pressure lowering properties. Moreover, anti-inflammatory drugs (mycophenolate mofetil) have been shown to decrease blood pressure in hypertensive patients or prevent its development in normotensive individuals. Further research is needed to evaluate whether drugs targeting hypertensive-linked proinflammatory cytokines, such as monoclonal antibodies, could become a new therapeutic option in treating arterial hypertension.
Collapse
|
38
|
Fisher CL, Demel SL. Nonsteroidal Anti-Inflammatory Drugs: A Potential Pharmacological Treatment for Intracranial Aneurysm. Cerebrovasc Dis Extra 2019; 9:31-45. [PMID: 31039577 PMCID: PMC7036563 DOI: 10.1159/000499077] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2018] [Accepted: 02/22/2019] [Indexed: 01/25/2023] Open
Abstract
Background Saccular intracranial aneurysms (IAs) are outpouchings of the vessel wall of intracranial arteries. Rupture of IAs results in subarachnoid hemorrhage which is associated with high morbidity and mortality. Surgical interventions, such as clipping and coiling, have associated risks. Currently, there are no proven pharmacological treatments to prevent the growth or rupture of IAs. Infiltration of proinflammatory cytokines in response to increased wall sheer stress is a hallmark of IA. Nonsteroidal anti-inflammatory drugs (NSAIDs) are being investigated as potential therapeutic agents for reduction in growth and/or prevention of IA through inhibition of inflammatory pathways. Summary This review will discuss the role of NSAIDs in attenuating the inflammation that drives IA progression and rupture. There are two main subtypes of NSAIDs, nonselective COX and selective COX-2 inhibitors, both of which have merit in treating IA. Evidence will be presented which shows that NSAIDs inhibit several key inflammatory mediators involved in IA progression including nuclear factor-κB, tumor necrosis factor-α, and matrix metalloproteinases. In addition, the role of NSAIDs in limiting inflammatory cell adhesion to endothelial cells and attenuating endothelial cell senescence will be discussed. Key Messages There is an abundance of basic science and preclinical data that support NSAIDs as a promising treatment for IA. Additionally, a combination treatment strategy of low-dose aspirin given concomitantly with a selective COX-2 inhibitor may result in a reduced side effect profile compared to aspirin or selective COX-2 inhibitor use alone. Several large clinical trials are currently planned to further investigate the efficacy of NSAIDs as an effective nonsurgical treatment for IAs.
Collapse
Affiliation(s)
- Courtney L Fisher
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, Michigan, USA,
| | - Stacie L Demel
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, Michigan, USA
| |
Collapse
|
39
|
Malik M, Britten J, Borahay M, Segars J, Catherino WH. Simvastatin, at clinically relevant concentrations, affects human uterine leiomyoma growth and extracellular matrix production. Fertil Steril 2019; 110:1398-1407.e1. [PMID: 30503138 DOI: 10.1016/j.fertnstert.2018.07.024] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2018] [Revised: 07/23/2018] [Accepted: 07/26/2018] [Indexed: 12/17/2022]
Abstract
OBJECTIVE To observe the antifibroid effects of therapeutic concentrations of simvastatin, which interferes with cholesterol biosynthesis, a known precursor of five major classes of steroid hormones, including progesterone and estrogen, which play a major role in the development and growth of uterine leiomyomas. DESIGN Two-dimensional and three-dimensional cell culture study of immortalized human leiomyoma and patient-matched myometrium cells treated with simvastatin. SETTING University laboratory. PATIENT(S) None. INTERVENTIONS(S) None. MAIN OUTCOME MEASURE(S) Cell proliferation, alteration in apoptotic signaling pathways, and extracellular matrix (ECM) protein production. RESULT(S) Simvastatin demonstrated a concentration-dependent antiproliferative effect on both the leiomyoma cells and the patient-matched myometrium cells, but a higher inhibitory effect at lower concentrations of simvastatin was observed in leiomyoma cells. Simvastatin also regulated leiomyoma cell apoptosis through a concentration-dependent increase in activity of caspase-3. Simvastatin significantly inhibited expression of major ECM proteins collagen I, collagen III, fibronectin, versican, and brevican in leiomyoma cells at concentrations as low as 10-9 mol/L within 48 hours of exposure. CONCLUSION(S) Simvastatin induces apoptosis in uterine leiomyoma cells at low concentrations, as evidenced by increased active caspase levels. Furthermore, inhibited production of the ECM proteins may lead to reduction in tumor size. Simvastatin may represent a novel therapeutic treatment strategy for uterine leiomyomas.
Collapse
Affiliation(s)
- Minnie Malik
- Department of Obstetrics and Gynecology, Uniformed Services University of the Health Sciences, Bethesda, Maryland
| | - Joy Britten
- Department of Obstetrics and Gynecology, Uniformed Services University of the Health Sciences, Bethesda, Maryland
| | - Mostafa Borahay
- Division of Reproductive Sciences and Women's Health Research, Department of Gynecology and Obstetrics, Johns Hopkins Medicine, Baltimore, Maryland
| | - James Segars
- Division of Reproductive Sciences and Women's Health Research, Department of Gynecology and Obstetrics, Johns Hopkins Medicine, Baltimore, Maryland
| | - William H Catherino
- Department of Obstetrics and Gynecology, Uniformed Services University of the Health Sciences, Bethesda, Maryland.
| |
Collapse
|
40
|
Hsu PL, Chen JS, Wang CY, Wu HL, Mo FE. Shear-Induced CCN1 Promotes Atheroprone Endothelial Phenotypes and Atherosclerosis. Circulation 2019; 139:2877-2891. [PMID: 30917686 DOI: 10.1161/circulationaha.118.033895] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND Atherosclerosis occurs preferentially at the blood vessels encountering blood flow turbulence. The matricellular protein CCN1 is induced in endothelial cells by disturbed flow, and is expressed in advanced atherosclerotic lesions in patients and in the Apoe-/- mouse model. The role of CCN1 in atherosclerosis remains undefined. METHODS To assess the function of CCN1 in vivo, knock-in mice carrying the integrin α6β1-binding-defective mutant allele Ccn1-dm on the Apoe-/- background were tested in an atherosclerosis model generated by carotid artery ligation. Additionally, CCN1-regulated functional phenotypes of human umbilical vein endothelial cells, or primary mouse aortic endothelial cells isolated from wild-type and Ccn1 dm/dm mice, were investigated in the in vitro shear stress experiments under unidirectional laminar shear stress (12 dyn/cm2) versus oscillatory shear stress (±5 dyn/cm2) conditions. RESULTS We found that Ccn1 expression was upregulated in the arterial endothelium 3 days after ligation before any detectable structural changes, and intensified with the progression of atherosclerotic lesions. Compared with Apoe-/- controls, Ccn1 dm/dm/ Apoe-/- mice were remarkably resistant to ligation-induced plaque formation (n=6). These mice exhibited lower oxidative stress, expression of endothelin-1 and monocyte chemoattractant protein-1, and monocyte homing. CCN1/α6β1 critically mediated flow-induced activation of the pleiotropic transcription factor nuclear factor-κB and therefore the induction of atheroprone gene expression in the mouse arterial endothelium after ligation (n=6), or in cultured human umbilical vein endothelial cells or primary mouse aortic endothelial cells exposed to oscillatory shear stress (n=3 in triplicate). Interestingly, the activation of nuclear factor-κB by CCN1/α6β1 signaling prompted more production of CCN1 and α6β1. Blocking CCN1-α6β1 binding by the Ccn1-dm mutation or by T1 peptide (derived from an α6β1-binding sequence of CCN1) disrupted the positive-feedback regulation between CCN1/α6β1 and nuclear factor-κB, and prevented flow-induced atheroprone phenotypic alterations in endothelial cells or atherosclerosis in mice. CONCLUSIONS These data demonstrate a causative role of CCN1 in atherosclerosis via modulating endothelial phenotypes. CCN1 binds to its receptor integrin α6β1 to activate nuclear factor-κB, thereby instigating a vicious circle to persistently promote atherogenesis. T1, a peptide antagonist selectively targeting CCN1-α6β1, can be further optimized for developing T1-mimetics to treat atherosclerosis.
Collapse
Affiliation(s)
- Pei-Ling Hsu
- Department of Cell Biology and Anatomy (P.-L.H., J.-S.C., C.-Y.W., F.-E M.), College of Medicine, National Cheng Kung University, Tainan, Taiwan.,Institute of Basic Medical Sciences (P.-L.H., H.-L.W., F.-E M.), College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Jheng-Sin Chen
- Department of Cell Biology and Anatomy (P.-L.H., J.-S.C., C.-Y.W., F.-E M.), College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Chin-Yung Wang
- Department of Cell Biology and Anatomy (P.-L.H., J.-S.C., C.-Y.W., F.-E M.), College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Hua-Lin Wu
- Institute of Basic Medical Sciences (P.-L.H., H.-L.W., F.-E M.), College of Medicine, National Cheng Kung University, Tainan, Taiwan.,Department of Biochemistry and Molecular Biology (H.-L.W.), College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Fan-E Mo
- Department of Cell Biology and Anatomy (P.-L.H., J.-S.C., C.-Y.W., F.-E M.), College of Medicine, National Cheng Kung University, Tainan, Taiwan.,Institute of Basic Medical Sciences (P.-L.H., H.-L.W., F.-E M.), College of Medicine, National Cheng Kung University, Tainan, Taiwan
| |
Collapse
|
41
|
Abstract
Current management of aortic aneurysms relies exclusively on prophylactic operative repair of larger aneurysms. Great potential exists for successful medical therapy that halts or reduces aneurysm progression and hence alleviates or postpones the need for surgical repair. Preclinical studies in the context of abdominal aortic aneurysm identified hundreds of candidate strategies for stabilization, and data from preoperative clinical intervention studies show that interventions in the pathways of the activated inflammatory and proteolytic cascades in enlarging abdominal aortic aneurysm are feasible. Similarly, the concept of pharmaceutical aorta stabilization in Marfan syndrome is supported by a wealth of promising studies in the murine models of Marfan syndrome-related aortapathy. Although some clinical studies report successful medical stabilization of growing aortic aneurysms and aortic root stabilization in Marfan syndrome, these claims are not consistently confirmed in larger and controlled studies. Consequently, no medical therapy can be recommended for the stabilization of aortic aneurysms. The discrepancy between preclinical successes and clinical trial failures implies shortcomings in the available models of aneurysm disease and perhaps incomplete understanding of the pathological processes involved in later stages of aortic aneurysm progression. Preclinical models more reflective of human pathophysiology, identification of biomarkers to predict severity of disease progression, and improved design of clinical trials may more rapidly advance the opportunities in this important field.
Collapse
Affiliation(s)
- Jan H. Lindeman
- Dept. Vascular Surgery, Leiden University Medical Center, The Netherlands
| | - Jon S. Matsumura
- Division of Vascular Surgery, School of Medicine and Public Health, University of Wisconsin, Madison, Wisconsin
| |
Collapse
|
42
|
Zhao J, Cheng Q, Liu Y, Yang G, Wang X. Atorvastatin alleviates early hypertensive renal damage in spontaneously hypertensive rats. Biomed Pharmacother 2019; 109:602-609. [DOI: 10.1016/j.biopha.2018.10.165] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2018] [Revised: 10/26/2018] [Accepted: 10/26/2018] [Indexed: 10/27/2022] Open
|
43
|
Abstract
Multiple in vitro and mice model studies suggest statins may attenuate dengue severity. However, little is known about statin use and dengue severity in adult dengue patients with hyperlipidemia. We conducted a retrospective cohort study from 2004–2008 and 2012–2013 in Tan Tock Seng Hospital, Singapore on adult dengue patients with hyperlipidemia, comparing those with and without statin usage at hospitalization in terms of primary outcome of dengue hemorrhagic fever (DHF) or shock syndrome (DSS), and severe dengue (SD). Of 13,975 subjects screened, 257 dengue patients were included; 191 (74.3%) were statin users and 66 (25.7%) were non-users. Compared with non-users, statin use was not associated with decreased risk of DHF/DSS (adjusted risk ratio [aRR] = 0.66, 95%confidence interval [CI]: 0.41–1.08, P = 0.10) and SD (aRR = 1.43, 95%CI: 0.84–2.43, P = 0.19). Therefore, statin usage had minimal effect on dengue severity in our study population in Singapore.
Collapse
|
44
|
Lee D, Joo HJ, Jung HW, Lim DS. Investigating potential mediator between statin and coronary artery calcification. PLoS One 2018; 13:e0203702. [PMID: 30226851 PMCID: PMC6143241 DOI: 10.1371/journal.pone.0203702] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2018] [Accepted: 08/25/2018] [Indexed: 12/22/2022] Open
Abstract
Statins are mainstay anti-lipidaemic treatments for preventing cardiovascular diseases but also known to increase coronary artery calcification (CAC). However, underlying relationship between statin and CAC is still unclear. This study explored the mediating role of five statin-related biochemical factors [i.e., low-density lipoprotein (LDL)-cholesterol, high-density lipoprotein (HDL)-cholesterol, triglyceride, glucose, and high sensitivity C-reactive protein levels]. Seoul Metabolic Syndrome cohort study includes 1370 participants suspected of metabolic syndrome. For causal mediation analysis, the dataset for 2016 including 847 participants with coronary computed tomography without any missing value were analysed using the Mediation package in R software. This study identified a causal mediation mechanism of HDL-cholesterol among the five biochemical factors. It implied that statin treatment increases the HDL-cholesterol level, leading to decreasing the probability of CAC score > 0. Estimated values of interest in HDL-cholesterol mediation were (1) average causal mediation effect, -0.011 with 95% CI [-0.025, -0.003], (2) average direct effect, 0.143 with 95% CI [0.074, 0.219], and total effect, 0.132 with 95% CI [0.063, 0.209]. Its mediation effect was maintained regardless of statin intensity. Sensitivity analysis also provided a robustness of the results under potential existence of a confounder between HDL-cholesterol and CAC. This study suggests a potential causal pathway between statin and CAC (the positive association of statin on CAC) through HDL-cholesterol as an inhibitor.
Collapse
Affiliation(s)
- Donghun Lee
- Korea University Business School, Seoul, Korea
| | - Hyung Joon Joo
- Department of Cardiology, Cardiovascular Center, Korea University Anam Hospital, Seoul, Korea
| | - Ho-Won Jung
- Korea University Business School, Seoul, Korea
| | - Do-Sun Lim
- Department of Cardiology, Cardiovascular Center, Korea University Anam Hospital, Seoul, Korea
| |
Collapse
|
45
|
Askarizadeh A, Butler AE, Badiee A, Sahebkar A. Liposomal nanocarriers for statins: A pharmacokinetic and pharmacodynamics appraisal. J Cell Physiol 2018; 234:1219-1229. [DOI: 10.1002/jcp.27121] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2018] [Accepted: 07/05/2018] [Indexed: 12/16/2022]
Affiliation(s)
- Anis Askarizadeh
- Nanotechnology Research Center Pharmaceutical Technology Institute, Mashhad University of Medical Sciences Mashhad Iran
| | | | - Ali Badiee
- Nanotechnology Research Center Pharmaceutical Technology Institute, Mashhad University of Medical Sciences Mashhad Iran
| | - Amirhossein Sahebkar
- Biotechnology Research Center Pharmaceutical Technology Institute, Mashhad University of Medical Sciences Mashhad Iran
- Neurogenic Inflammation Research Center Mashhad University of Medical Sciences Mashhad Iran
- School of Pharmacy, Mashhad University of Medical Sciences Mashhad Iran
| |
Collapse
|
46
|
Sequeira Gross T, Naito S, Neumann N, Petersen J, Kuntze T, Reichenspurner H, von Kodolitsch Y, Girdauskas E. Does statin therapy impact the proximal aortopathy in aortic valve disease? QJM 2018; 111:623-628. [PMID: 29917097 DOI: 10.1093/qjmed/hcy129] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/13/2018] [Indexed: 11/14/2022] Open
Abstract
BACKGROUND Studies have demonstrated that statin therapy decreases the growth rate of abdominal aneurysms. However, the effect of statin therapy on the proximal aortic disease has not been sufficiently elucidated. AIM We aimed to analyse the association between statin treatment and the severity of proximal aortopathy in patients with aortic valve disease. DESIGN Cross-sectional study. METHODS We prospectively identified 458 patients who were referred for aortic valve surgery from 2008 to 2014. Pre-operative measurement of the proximal aorta was performed by TEE, CT or MRT scan. Data of dyslipidemia treatment was obtained by questionnaire. RESULTS The mean ascending aortic diameter in the whole study population was comparable in patients with vs. without statin therapy (i.e. 42.7 mm vs. 43.6 mm, P = 0.46). Logistic regression analysis showed no significant association between statin therapy and proximal aortopathy ≥ 40 mm in the whole study group (OR = 0.69, P = 0.10). For the BAV sub-group, HDL (OR = 0.54, P = 0.038) and cholesterol levels (OR = 2.00, P = 0.038) were found significantly associated with the proximal aortic disease. In the BAV cohort, the statin users with target HDL levels presented a significantly smaller proximal aortic diameter (40.1 mm vs. 46 mm, P = 0.02). CONCLUSION Pre-operative statin therapy demonstrated no significant association with the expression of proximal aortopathy. However, more than 40% of the statin users presented uncontrolled lipid levels at the time of the study. In the BAV sub-group, the statins users with target HDL levels showed a significantly smaller ascending aorta diameter. Target HDL and cholesterol levels were strongly associated with proximal aortic dilation in BAV patients.
Collapse
Affiliation(s)
- T Sequeira Gross
- From the Department of Cardiovascular Surgery, University Heart Center Hamburg, Martinistraße 52, Hamburg, Germany
| | - S Naito
- From the Department of Cardiovascular Surgery, University Heart Center Hamburg, Martinistraße 52, Hamburg, Germany
| | - N Neumann
- From the Department of Cardiovascular Surgery, University Heart Center Hamburg, Martinistraße 52, Hamburg, Germany
| | - J Petersen
- From the Department of Cardiovascular Surgery, University Heart Center Hamburg, Martinistraße 52, Hamburg, Germany
| | - T Kuntze
- Department of Cardiac Surgery, Central Hospital Bad Berka, Robert Koch Alle 9, Bad Berka, Germany
| | - H Reichenspurner
- From the Department of Cardiovascular Surgery, University Heart Center Hamburg, Martinistraße 52, Hamburg, Germany
| | - Y von Kodolitsch
- Department of Cardiology, University Heart Center Hamburg, Martinistraße 52, Hamburg, Germany
| | - E Girdauskas
- From the Department of Cardiovascular Surgery, University Heart Center Hamburg, Martinistraße 52, Hamburg, Germany
| |
Collapse
|
47
|
Rossignoli A, Vorkapic E, Wanhainen A, Länne T, Skogberg J, Folestad E, Wågsäter D. Plasma cholesterol lowering in an AngII‑infused atherosclerotic mouse model with moderate hypercholesterolemia. Int J Mol Med 2018; 42:471-478. [PMID: 29658561 DOI: 10.3892/ijmm.2018.3619] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2017] [Accepted: 02/22/2018] [Indexed: 11/06/2022] Open
Abstract
Atherosclerosis is the main underlying causes of cardiovascular disease. There is a well‑established association between high blood cholesterol levels and the extent of atherosclerosis. Furthermore, atherosclerosis has been proposed to augment abdominal aortic aneurysm (AAA) formation. As patients with AAA often have parallel atherosclerotic disease and are therefore often on cholesterol‑lowering therapy, it is not possible to fully address the independent effects of plasma cholesterol lowering (PCL) treatment on AAA. The present study investigated the effect of angiotensin II (AngII)‑infusion in modestly hypercholesterolemic Ldlr‑/‑Apob100/100Mttpflox/floxMx1‑Cre mice with or without PCL treatment on a morphological and molecular level, in terms of atherosclerosis and AAA development. AngII infusion in the study mice resulted in an increased atherosclerotic lesion area and increased infiltration of inflammatory leukocytes, which was not observed in mice with PCL induced prior to AngII infusion. This suggested that AngII infusion in this mouse model induced atherosclerosis development, and that plasma cholesterol levels represent a controlling factor. Furthermore, AngII infusion in Ldlr‑/‑Apob100/100Mttpflox/floxMx1‑Cre mice caused a modest aneurysmal phenotype, and no differences in AAA development were observed between the different study groups. However, the fact that modest hypercholesterolemic mice did not develop AAA in a classical aneurysmal model indicated that plasma cholesterol levels are important for disease development.
Collapse
Affiliation(s)
- Aránzazu Rossignoli
- Division of Vascular Biology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, 17177 Stockholm, Sweden
| | - Emina Vorkapic
- Division of Drug Research, Department of Medical and Health Sciences, Linköping University, 58183 Linköping, Sweden
| | - Anders Wanhainen
- Department of Surgical Sciences, Section of Vascular Surgery, Uppsala University, 75185 Uppsala, Sweden
| | - Toste Länne
- Division of Cardiovascular Medicine, Department of Medical and Health Sciences, Linköping University, 58183 Linköping, Sweden
| | - Josefin Skogberg
- Division of Vascular Biology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, 17177 Stockholm, Sweden
| | - Erika Folestad
- Division of Vascular Biology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, 17177 Stockholm, Sweden
| | - Dick Wågsäter
- Division of Drug Research, Department of Medical and Health Sciences, Linköping University, 58183 Linköping, Sweden
| |
Collapse
|
48
|
Kozijn AE, Gierman LM, van der Ham F, Mulder P, Morrison MC, Kühnast S, van der Heijden RA, Stavro PM, van Koppen A, Pieterman EJ, van den Hoek AM, Kleemann R, Princen HMG, Mastbergen SC, Lafeber FPJG, Zuurmond AM, Bobeldijk I, Weinans H, Stoop R. Variable cartilage degradation in mice with diet-induced metabolic dysfunction: food for thought. Osteoarthritis Cartilage 2018; 26:95-107. [PMID: 29074298 DOI: 10.1016/j.joca.2017.10.010] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/01/2017] [Revised: 10/03/2017] [Accepted: 10/10/2017] [Indexed: 02/02/2023]
Abstract
OBJECTIVE Human cohort studies have demonstrated a role for systemic metabolic dysfunction in osteoarthritis (OA) pathogenesis in obese patients. To explore the mechanisms underlying this metabolic phenotype of OA, we examined cartilage degradation in the knees of mice from different genetic backgrounds in which a metabolic phenotype was established by various dietary approaches. DESIGN Wild-type C57BL/6J mice and genetically modified mice (hCRP, LDLr-/-. Leiden and ApoE*3Leiden.CETP mice) based on C57BL/6J background were used to investigate the contribution of inflammation and altered lipoprotein handling on diet-induced cartilage degradation. High-caloric diets of different macronutrient composition (i.e., high-carbohydrate or high-fat) were given in regimens of varying duration to induce a metabolic phenotype with aggravated cartilage degradation relative to controls. RESULTS Metabolic phenotypes were confirmed in all studies as mice developed obesity, hypercholesteremia, glucose intolerance and/or insulin resistance. Aggravated cartilage degradation was only observed in two out of the twelve experimental setups, specifically in long-term studies in male hCRP and female ApoE*3Leiden.CETP mice. C57BL/6J and LDLr-/-. Leiden mice did not develop HFD-induced OA under the conditions studied. Osteophyte formation and synovitis scores showed variable results between studies, but also between strains and gender. CONCLUSIONS Long-term feeding of high-caloric diets consistently induced a metabolic phenotype in various C57BL/6J (-based) mouse strains. In contrast, the induction of articular cartilage degradation proved variable, which suggests that an additional trigger might be necessary to accelerate diet-induced OA progression. Gender and genetic modifications that result in a humanized pro-inflammatory state (human CRP) or lipoprotein metabolism (human-E3L.CETP) were identified as important contributing factors.
Collapse
Affiliation(s)
- A E Kozijn
- Metabolic Health Research, TNO, Leiden, The Netherlands; Department of Orthopaedics, University Medical Center (UMC) Utrecht, Utrecht University, Utrecht, The Netherlands; Department of Rheumatology & Clinical Immunology, UMC Utrecht, Utrecht University, Utrecht, The Netherlands
| | - L M Gierman
- Metabolic Health Research, TNO, Leiden, The Netherlands
| | - F van der Ham
- Metabolic Health Research, TNO, Leiden, The Netherlands
| | - P Mulder
- Metabolic Health Research, TNO, Leiden, The Netherlands
| | - M C Morrison
- Metabolic Health Research, TNO, Leiden, The Netherlands
| | - S Kühnast
- Metabolic Health Research, TNO, Leiden, The Netherlands
| | - R A van der Heijden
- Department of Pathology and Medical Biology, UMC Groningen, Groningen, The Netherlands
| | - P M Stavro
- Bunge North America, Saint Louis, United States
| | - A van Koppen
- Metabolic Health Research, TNO, Leiden, The Netherlands
| | - E J Pieterman
- Metabolic Health Research, TNO, Leiden, The Netherlands
| | | | - R Kleemann
- Metabolic Health Research, TNO, Leiden, The Netherlands
| | - H M G Princen
- Metabolic Health Research, TNO, Leiden, The Netherlands
| | - S C Mastbergen
- Department of Rheumatology & Clinical Immunology, UMC Utrecht, Utrecht University, Utrecht, The Netherlands
| | - F P J G Lafeber
- Department of Rheumatology & Clinical Immunology, UMC Utrecht, Utrecht University, Utrecht, The Netherlands
| | - A-M Zuurmond
- Metabolic Health Research, TNO, Leiden, The Netherlands
| | - I Bobeldijk
- Metabolic Health Research, TNO, Leiden, The Netherlands
| | - H Weinans
- Department of Orthopaedics, University Medical Center (UMC) Utrecht, Utrecht University, Utrecht, The Netherlands; Department of Rheumatology & Clinical Immunology, UMC Utrecht, Utrecht University, Utrecht, The Netherlands; Department of Biomechanical Engineering, Delft University of Technology, Delft, The Netherlands
| | - R Stoop
- Metabolic Health Research, TNO, Leiden, The Netherlands.
| |
Collapse
|
49
|
Yu P, Xiong T, Tenedero CB, Lebeau P, Ni R, MacDonald ME, Gross PL, Austin RC, Trigatti BL. Rosuvastatin Reduces Aortic Sinus and Coronary Artery Atherosclerosis in SR-B1 (Scavenger Receptor Class B Type 1)/ApoE (Apolipoprotein E) Double Knockout Mice Independently of Plasma Cholesterol Lowering. Arterioscler Thromb Vasc Biol 2017; 38:26-39. [PMID: 29162602 PMCID: PMC5757666 DOI: 10.1161/atvbaha.117.305140] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2017] [Accepted: 11/08/2017] [Indexed: 11/16/2022]
Abstract
Supplemental Digital Content is available in the text. Objective— Rosuvastatin has been widely used in the primary and secondary prevention of coronary heart disease. However, its antiatherosclerotic properties have not been tested in a mouse model that could mimic human coronary heart disease. The present study was designed to test the effects of rosuvastatin on coronary artery atherosclerosis and myocardial fibrosis in SR-B1 (scavenger receptor class B type 1) and apoE (apolipoprotein E) double knockout mice. Approach and Results— Three-week-old SR-B1−/−/apoE−/− mice were injected daily with 10 mg/kg of rosuvastatin for 2 weeks. Compared with saline-treated mice, rosuvastatin-treated mice showed increased levels of hepatic PCSK9 (proprotein convertase subtilisin/kexin type-9) and LDLR (low-density lipoprotein receptor) message, increased plasma PCSK9 protein but decreased levels of hepatic LDLR protein and increased plasma total cholesterol associated with apoB (apolipoprotein B) 48-containing lipoproteins. In spite of this, rosuvastatin treatment was associated with decreased atherosclerosis in both the aortic sinus and coronary arteries and reduced platelet accumulation in atherosclerotic coronary arteries. Cardiac fibrosis and cardiomegaly were also attenuated in rosuvastatin-treated SR-B1−/−/apoE−/− mice. Two-week treatment with rosuvastatin resulted in significant decreases in markers of oxidized phospholipids in atherosclerotic plaques. In vitro analysis showed that incubation of bone marrow-derived macrophages with rosuvastatin substantially downregulated cluster of differentiation (CD)36 and inhibited oxidized LDL-induced foam cell formation. Conclusions— Rosuvastatin protected SR-B1−/−/apoE−/− mice against atherosclerosis and platelet accumulation in coronary arteries and attenuated myocardial fibrosis and cardiomegaly, despite increased plasma total cholesterol. The ability of rosuvastatin to reduce oxidized phospholipids in atherosclerotic plaques and inhibit macrophage foam cell formation may have contributed to this protection.
Collapse
Affiliation(s)
- Pei Yu
- From the Thrombosis and Atherosclerosis Research Institute, McMaster University and Hamilton Health Sciences (P.Y., T.X., C.B.T., R.N., M.E.M., P.L.G., R.C.A., B.L.T.), St. Joseph's Hamilton Healthcare and Hamilton Center for Kidney Research (P.L., R.C.A.), Department of Biochemistry and Biomedical Sciences (P.Y., T.X., C.B.T., M.E.M., B.L.T.), and Department of Medicine (P.L., R.N., P.L.G., R.C.A.), McMaster University, Hamilton, ON, Canada
| | - Ting Xiong
- From the Thrombosis and Atherosclerosis Research Institute, McMaster University and Hamilton Health Sciences (P.Y., T.X., C.B.T., R.N., M.E.M., P.L.G., R.C.A., B.L.T.), St. Joseph's Hamilton Healthcare and Hamilton Center for Kidney Research (P.L., R.C.A.), Department of Biochemistry and Biomedical Sciences (P.Y., T.X., C.B.T., M.E.M., B.L.T.), and Department of Medicine (P.L., R.N., P.L.G., R.C.A.), McMaster University, Hamilton, ON, Canada
| | - Christine B Tenedero
- From the Thrombosis and Atherosclerosis Research Institute, McMaster University and Hamilton Health Sciences (P.Y., T.X., C.B.T., R.N., M.E.M., P.L.G., R.C.A., B.L.T.), St. Joseph's Hamilton Healthcare and Hamilton Center for Kidney Research (P.L., R.C.A.), Department of Biochemistry and Biomedical Sciences (P.Y., T.X., C.B.T., M.E.M., B.L.T.), and Department of Medicine (P.L., R.N., P.L.G., R.C.A.), McMaster University, Hamilton, ON, Canada
| | - Paul Lebeau
- From the Thrombosis and Atherosclerosis Research Institute, McMaster University and Hamilton Health Sciences (P.Y., T.X., C.B.T., R.N., M.E.M., P.L.G., R.C.A., B.L.T.), St. Joseph's Hamilton Healthcare and Hamilton Center for Kidney Research (P.L., R.C.A.), Department of Biochemistry and Biomedical Sciences (P.Y., T.X., C.B.T., M.E.M., B.L.T.), and Department of Medicine (P.L., R.N., P.L.G., R.C.A.), McMaster University, Hamilton, ON, Canada
| | - Ran Ni
- From the Thrombosis and Atherosclerosis Research Institute, McMaster University and Hamilton Health Sciences (P.Y., T.X., C.B.T., R.N., M.E.M., P.L.G., R.C.A., B.L.T.), St. Joseph's Hamilton Healthcare and Hamilton Center for Kidney Research (P.L., R.C.A.), Department of Biochemistry and Biomedical Sciences (P.Y., T.X., C.B.T., M.E.M., B.L.T.), and Department of Medicine (P.L., R.N., P.L.G., R.C.A.), McMaster University, Hamilton, ON, Canada
| | - Melissa E MacDonald
- From the Thrombosis and Atherosclerosis Research Institute, McMaster University and Hamilton Health Sciences (P.Y., T.X., C.B.T., R.N., M.E.M., P.L.G., R.C.A., B.L.T.), St. Joseph's Hamilton Healthcare and Hamilton Center for Kidney Research (P.L., R.C.A.), Department of Biochemistry and Biomedical Sciences (P.Y., T.X., C.B.T., M.E.M., B.L.T.), and Department of Medicine (P.L., R.N., P.L.G., R.C.A.), McMaster University, Hamilton, ON, Canada
| | - Peter L Gross
- From the Thrombosis and Atherosclerosis Research Institute, McMaster University and Hamilton Health Sciences (P.Y., T.X., C.B.T., R.N., M.E.M., P.L.G., R.C.A., B.L.T.), St. Joseph's Hamilton Healthcare and Hamilton Center for Kidney Research (P.L., R.C.A.), Department of Biochemistry and Biomedical Sciences (P.Y., T.X., C.B.T., M.E.M., B.L.T.), and Department of Medicine (P.L., R.N., P.L.G., R.C.A.), McMaster University, Hamilton, ON, Canada
| | - Richard C Austin
- From the Thrombosis and Atherosclerosis Research Institute, McMaster University and Hamilton Health Sciences (P.Y., T.X., C.B.T., R.N., M.E.M., P.L.G., R.C.A., B.L.T.), St. Joseph's Hamilton Healthcare and Hamilton Center for Kidney Research (P.L., R.C.A.), Department of Biochemistry and Biomedical Sciences (P.Y., T.X., C.B.T., M.E.M., B.L.T.), and Department of Medicine (P.L., R.N., P.L.G., R.C.A.), McMaster University, Hamilton, ON, Canada
| | - Bernardo L Trigatti
- From the Thrombosis and Atherosclerosis Research Institute, McMaster University and Hamilton Health Sciences (P.Y., T.X., C.B.T., R.N., M.E.M., P.L.G., R.C.A., B.L.T.), St. Joseph's Hamilton Healthcare and Hamilton Center for Kidney Research (P.L., R.C.A.), Department of Biochemistry and Biomedical Sciences (P.Y., T.X., C.B.T., M.E.M., B.L.T.), and Department of Medicine (P.L., R.N., P.L.G., R.C.A.), McMaster University, Hamilton, ON, Canada.
| |
Collapse
|
50
|
Groeneveld ME, van der Reijden JJ, Tangelder GJ, Westin LC, Renwarin L, Musters RJP, Wisselink W, Yeung KK. Peroxynitrite Footprint in Circulating Neutrophils of Abdominal Aortic Aneurysm Patients is Lower in Statin than in Non-statin Users. Eur J Vasc Endovasc Surg 2017; 54:331-339. [PMID: 28712812 DOI: 10.1016/j.ejvs.2017.06.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2016] [Accepted: 06/02/2017] [Indexed: 01/08/2023]
Abstract
OBJECTIVES Extensive reactive oxygen and nitrogen species (also reactive species) production is a mechanism involved in abdominal aortic aneurysm (AAA) development. White blood cells (WBCs) are a known source of reactive species. Their production may be decreased by statins, thereby reducing the AAA growth rate. Reactive species production in circulating WBCs of AAA patients and the effect of statins on their production was investigated. METHODS This observational study investigated reactive species production in vivo and ex vivo in circulating WBCs of AAA patients, using venous blood from patients prior to elective AAA repair (n = 34; 18 statin users) and from healthy volunteers (n = 10). Reactive species production was quantified in circulating WBCs using immunofluorescence microscopy: nitrotyrosine (footprint of peroxynitrite, a potent reactive nitrogen species) in snap frozen blood smears; mitochondrial superoxide and cytoplasmic hydrogen peroxide (both reactive oxygen species) by live cell imaging. Neutrophils, lymphocytes, and monocytes were examined individually. RESULTS In AAA patients using statins, the median nitrotyrosine level in neutrophils was 646 (range 422-2059), in lymphocytes 125 (range 74-343), and in monocytes 586 (range 291-663). Median levels in AAA patients not using statins were for neutrophils 928 (range 552-2095, p = .03), lymphocytes 156 (101-273, NS), and for monocytes 536 (range 535-1635, NS). The statin dose tended to correlate negatively with nitrotyrosine in neutrophils (Rs -0.32, p = .06). The median levels in controls were lower for neutrophils 466 (range 340-820, p < .01) and for monocytes 191 (range 102-386, p = .03), but similar for lymphocytes 99 (range 82-246) when compared to the AAA patients. There were no differences in mitochondrial superoxide and cytoplasmic hydrogen peroxide between statin and non-statin users within AAA patients. CONCLUSIONS It was found that the peroxynitrite footprint in circulating neutrophils and monocytes of AAA patients is higher than in controls. AAA patients treated with statins had a lower peroxynitrite footprint in neutrophils than non-statin users.
Collapse
Affiliation(s)
- M E Groeneveld
- Department of Vascular Surgery, ICaR-VU, VU University Medical Center (VUmc), Amsterdam, The Netherlands; Department of Physiology, ICaR-VU, VU University Medical Center (VUmc), Amsterdam, The Netherlands
| | | | - G J Tangelder
- Department of Physiology, ICaR-VU, VU University Medical Center (VUmc), Amsterdam, The Netherlands
| | - L C Westin
- Center for Digestive Diseases, Karolinska University Hospital, Karolinska, Sweden
| | - L Renwarin
- Medical Department, Royal Netherlands Navy, Eindhoven, The Netherlands
| | - R J P Musters
- Department of Physiology, ICaR-VU, VU University Medical Center (VUmc), Amsterdam, The Netherlands
| | - W Wisselink
- Department of Vascular Surgery, ICaR-VU, VU University Medical Center (VUmc), Amsterdam, The Netherlands
| | - K K Yeung
- Department of Vascular Surgery, ICaR-VU, VU University Medical Center (VUmc), Amsterdam, The Netherlands; Department of Physiology, ICaR-VU, VU University Medical Center (VUmc), Amsterdam, The Netherlands.
| |
Collapse
|