1
|
Mao J, Wu C, Zheng L, Li Y, Yang R, Yuan P, Jiang J, Li C, Zhou X. Advances in stimulus-responsive nanomedicine for treatment and diagnosis of atherosclerosis. Colloids Surf B Biointerfaces 2025; 245:114298. [PMID: 39378703 DOI: 10.1016/j.colsurfb.2024.114298] [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: 08/20/2024] [Revised: 09/22/2024] [Accepted: 10/04/2024] [Indexed: 10/10/2024]
Abstract
Atherosclerosis (AS), an inflammatory cardiovascular disease driven by lipid deposition, presents global prevalence with high mortality. Effective anti-inflammatory or lipid removal is a promising strategy. However, current conventional drug delivery methods may face challenges in targeting disease sites and are deficient in the treatment of AS because of the nonspecific tissue distribution and uncontrollable release of the drug. In contrast, stimulus-responsive nanodrug delivery systems (NDDSs) can respond to stimulation and achieve controlled drug release rates at specific disease sites owing to the abnormal pathological microenvironment in plaques with low pH, excessive reactive oxygen species (ROS) and enzymes, and high shear stress. As a consequence, the efficacy of treatment is improved, and adverse reactions are reduced. On the other hand, NDDSs can combine exogenous stimulus responses (photothermal, ultrasound, etc.) to precisely control their function in time and space. This review for the first time focuses on the application of stimulus-responsive NDDSs in the treatment and diagnosis of AS in the last five years. In addition, its pivotal challenges and prospects are emphasized, aiming to facilitate its application for AS.
Collapse
Affiliation(s)
- Jingying Mao
- Basic Medicine Research Innovation Center for Cardiometabolic Diseases, Ministry of Education, Luzhou, Sichuan 646000, China
| | - Chengxi Wu
- Basic Medicine Research Innovation Center for Cardiometabolic Diseases, Ministry of Education, Luzhou, Sichuan 646000, China; Department of Thyroid and Vascular Surgery, the Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Lixin Zheng
- Basic Medicine Research Innovation Center for Cardiometabolic Diseases, Ministry of Education, Luzhou, Sichuan 646000, China; Department of Thyroid and Vascular Surgery, the Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Yaoyao Li
- Basic Medicine Research Innovation Center for Cardiometabolic Diseases, Ministry of Education, Luzhou, Sichuan 646000, China; Department of Thyroid and Vascular Surgery, the Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Ronghao Yang
- Basic Medicine Research Innovation Center for Cardiometabolic Diseases, Ministry of Education, Luzhou, Sichuan 646000, China; Department of Thyroid and Vascular Surgery, the Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Ping Yuan
- Department of Neurology, the Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Jun Jiang
- Department of Thyroid and Vascular Surgery, the Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Chunhong Li
- Department of Pharmaceutical Sciences, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646000, China.
| | - Xiangyu Zhou
- Basic Medicine Research Innovation Center for Cardiometabolic Diseases, Ministry of Education, Luzhou, Sichuan 646000, China; Department of Thyroid and Vascular Surgery, the Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China.
| |
Collapse
|
2
|
Lusta KA, Churov AV, Beloyartsev DF, Golovyuk AL, Lee AA, Sukhorukov VN, Orekhov AN. The two coin sides of bacterial extracellular membrane nanovesicles: atherosclerosis trigger or remedy. DISCOVER NANO 2024; 19:179. [PMID: 39532781 PMCID: PMC11557815 DOI: 10.1186/s11671-024-04149-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Accepted: 11/04/2024] [Indexed: 11/16/2024]
Abstract
Among the numerous driving forces that cause the atherosclerotic cardiovascular disease (ASCVD), pathogenic bacterial extracellular membrane nanovesicles (BEMNs) containing toxins and virulence factors appear to be the key trigger of inflammation and atherogenesis, the major processes involved in the pathogenesis of ASCVD. Since BEMNs are the carriers of nanosized biomolecules to distant sites, they are now being considered as a novel drug delivery system. Nowadays, many therapeutic strategies are used to treat ASCVD. However, the conventional anti-atherosclerotic therapies are not effective enough. This primarily due to the inefficiency of non-targeted drug delivery systems to tissue affected areas, which, in turn, leads to numerous side effects, as well as faulty pharmacokinetics. In this regard, nanomedicine methods using nanoparticles (NPs) as targeted drug delivery vehicles proved to be extremely useful. Bioengineered BEMNs equipped with disease-specific ligand moieties and loaded with corresponding drugs represent a promising tool in nanomedicine, which can be used as a novel drug delivery system for a successful therapy of ASCVD. In this review, we outline the involvement of pathogenic BEMNs in the triggering of ASCVD, the conventional therapeutic strategies for the treatment of ASCVD, and the recent trends in nanomedicine using BEMNs and NPs as a vehicle for targeted drug delivery.
Collapse
Affiliation(s)
- Konstantin A Lusta
- Institute for Atherosclerosis Research, Ltd, Osennyaya Street 4-1-207, Moscow, Russia, 121609.
| | - Alexey V Churov
- Institute on Aging Research, Russian Gerontology Clinical Research Center, Pirogov Russian National Research Medical University, Moscow, Russia, 129226
- Institute of General Pathology and Pathophysiology, 8 Baltiiskaya Street, Moscow, Russia, 125315
| | - Dmitry F Beloyartsev
- Vascular Surgery Department, A.V. Vishnevsky National Medical Research Center of Surgery, 27 Bolshaya Serpukhovskaya Street, Moscow, Russia, 117997
| | - Alexander L Golovyuk
- Vascular Surgery Department, A.V. Vishnevsky National Medical Research Center of Surgery, 27 Bolshaya Serpukhovskaya Street, Moscow, Russia, 117997
| | - Arthur A Lee
- Insitute of Human Morphology, Petrovsky Russian National Center of Surgery, 2 Abrikosovsky Lane, Moscow, Russia, 119991
| | - Vasily N Sukhorukov
- Insitute of Human Morphology, Petrovsky Russian National Center of Surgery, 2 Abrikosovsky Lane, Moscow, Russia, 119991
- Institute of General Pathology and Pathophysiology, 8 Baltiiskaya Street, Moscow, Russia, 125315
| | - Alexander N Orekhov
- Insitute of Human Morphology, Petrovsky Russian National Center of Surgery, 2 Abrikosovsky Lane, Moscow, Russia, 119991
- Institute of General Pathology and Pathophysiology, 8 Baltiiskaya Street, Moscow, Russia, 125315
| |
Collapse
|
3
|
Abduzhamalova NM, Mamedov MN. Prospects for the use of anti-inflammatory therapy in patients with coronary artery disease. RATIONAL PHARMACOTHERAPY IN CARDIOLOGY 2024; 20:550-558. [DOI: 10.20996/1819-6446-2024-3105] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2025] Open
Abstract
Coronary atherosclerosis is the primary cause of coronary artery disease, one of the world’s greatest causes of death. Inflammation plays a major role in atherosclerosis formation and progression. Cardiovascular disease risk factors such as dyslipidemia, hyperglycemia, smoking, hypertension and others contribute to endothelial injury. This sets off a cascade of inflammatory reactions accompanied by the release of cytokines and other inflammatory mediators, ultimately leading to rupture or erosion of the atherosclerotic plaque and atherothrombosis. Large studies such as CANTOS (Canakinumab Anti-Inflammatory Thrombosis Outcomes Study); COLCOT (Colchicine Cardiovascular Outcomes Trial) and LoDoCo2 (Low Dose Colchicine 2) demonstrated that the use of anti-inflammatory drugs improves the prognosis of patients with coronary atherosclerosis. At present, the most convincing evidence of the effectiveness of anti-inflammatory therapy was obtained only for colchicine and canakinumab. However, despite these promising results, there are still many issues to be addressed. Firstly, more research is needed to determine the optimal dosage and duration of these drugs. Secondly, the safety of their prolonged use should be carefully assessed, especially in the context of possible side effects. For example, anti-inflammatory drugs can potentially pose infectious risks that require special monitoring and follow-up. The review presents current views on the possibilities immunomodulatory drugs using in the complex treatment of atherosclerosis.
Collapse
Affiliation(s)
| | - M. N. Mamedov
- National Medical Research Center for Therapy and Preventive Medicine
| |
Collapse
|
4
|
Martí-Carvajal AJ, Gemmato-Valecillos MA, Monge Martín D, Dayer M, Alegría-Barrero E, De Sanctis JB, Parise Vasco JM, Riera Lizardo RJ, Nicola S, Martí-Amarista CE, Correa-Pérez A. Interleukin-receptor antagonist and tumour necrosis factor inhibitors for the primary and secondary prevention of atherosclerotic cardiovascular diseases. Cochrane Database Syst Rev 2024; 9:CD014741. [PMID: 39297531 PMCID: PMC11411914 DOI: 10.1002/14651858.cd014741.pub2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 09/21/2024]
Abstract
BACKGROUND Atherosclerotic cardiovascular disease (ACVD) is worsened by chronic inflammatory diseases. Interleukin receptor antagonists (IL-RAs) and tumour necrosis factor-alpha (TNF) inhibitors have been studied to see if they can prevent cardiovascular events. OBJECTIVES The purpose of this study was to assess the clinical benefits and harms of IL-RAs and TNF inhibitors in the primary and secondary prevention of ACVD. SEARCH METHODS The Cochrane Heart Specialised Register, the Cochrane Central Register of Controlled Trials (CENTRAL), Ovid MEDLINE (including In-Process & Other Non-Indexed Citations), Ovid Embase, EBSCO CINAHL plus, and clinical trial registries for ongoing and unpublished studies were searched in February 2024. The reference lists of relevant studies, reviews, meta-analyses and health technology reports were searched to identify additional studies. No limitations on language, date of publication or study type were set. SELECTION CRITERIA RCTs that recruited people with and without pre-existing ACVD, comparing IL-RAs or TNF inhibitors versus placebo or usual care, were selected. The primary outcomes considered were all-cause mortality, myocardial infarction, unstable angina, and adverse events. DATA COLLECTION AND ANALYSIS Two or more review authors, working independently at each step, selected studies, extracted data, assessed the risk of bias and used GRADE to judge the certainty of evidence. MAIN RESULTS We included 58 RCTs (22,053 participants; 21,308 analysed), comparing medication efficacy with placebo or usual care. Thirty-four trials focused on primary prevention and 24 on secondary prevention. The interventions included IL-1 RAs (anakinra, canakinumab), IL-6 RA (tocilizumab), TNF-inhibitors (etanercept, infliximab) compared with placebo or usual care. The certainty of evidence was low to very low due to biases and imprecision; all trials had a high risk of bias. Primary prevention: IL-1 RAs The evidence is very uncertain about the effects of the intervention on all-cause mortality(RR 0.33, 95% CI 0.01 to 7.58, 1 trial), myocardial infarction (RR 0.71, 95% CI 0.04 to 12.48, I² = 39%, 2 trials), unstable angina (RR 0.24, 95% CI 0.03 to 2.11, I² = 0%, 2 trials), stroke (RR 2.42, 95% CI 0.12 to 50.15; 1 trial), adverse events (RR 0.85, 95% CI 0.59 to 1.22, I² = 54%, 3 trials), or infection (rate ratio 0.84, 95% 0.55 to 1.29, I² = 0%, 4 trials). Evidence is very uncertain about whether anakinra and cankinumab may reduce heart failure (RR 0.21, 95% CI 0.05 to 0.94, I² = 0%, 3 trials). Peripheral vascular disease (PVD) was not reported as an outcome. IL-6 RAs The evidence is very uncertain about the effects of the intervention on all-cause mortality (RR 0.68, 95% CI 0.12 to 3.74, I² = 30%, 3 trials), myocardial infarction (RR 0.27, 95% CI 0.04 to1.68, I² = 0%, 3 trials), heart failure (RR 1.02, 95% CI 0.11 to 9.63, I² = 0%, 2 trials), PVD (RR 2.94, 95% CI 0.12 to 71.47, 1 trial), stroke (RR 0.34, 95% CI 0.01 to 8.14, 1 trial), or any infection (rate ratio 1.10, 95% CI: 0.88 to 1.37, I2 = 18%, 5 trials). Adverse events may increase (RR 1.13, 95% CI 1.04 to 1.23, I² = 33%, 5 trials). No trial assessed unstable angina. TNF inhibitors The evidence is very uncertain about the effects of the intervention on all-cause mortality (RR 1.78, 95% CI 0.63 to 4.99, I² = 10%, 3 trials), myocardial infarction (RR 2.61, 95% CI 0.11 to 62.26, 1 trial), stroke (RR 0.46, 95% CI 0.08 to 2.80, I² = 0%; 3 trials), heart failure (RR 0.85, 95% CI 0.06 to 12.76, 1 trial). Adverse events may increase (RR 1.13, 95% CI 1.01 to 1.25, I² = 51%, 13 trials). No trial assessed unstable angina or PVD. Secondary prevention: IL-1 RAs The evidence is very uncertain about the effects of the intervention on all-cause mortality (RR 0.94, 95% CI 0.84 to 1.06, I² = 0%, 8 trials), unstable angina (RR 0.88, 95% CI 0.65 to 1.19, I² = 0%, 3 trials), PVD (RR 0.85, 95% CI 0.19 to 3.73, I² = 38%, 3 trials), stroke (RR 0.94, 95% CI 0.74 to 1.2, I² = 0%; 7 trials), heart failure (RR 0.91, 95% 0.5 to 1.65, I² = 0%; 7 trials), or adverse events (RR 0.92, 95% CI 0.78 to 1.09, I² = 3%, 4 trials). There may be little to no difference between the groups in myocardial infarction (RR 0.88, 95% CI 0.0.75 to 1.04, I² = 0%, 6 trials). IL6-RAs The evidence is very uncertain about the effects of the intervention on all-cause mortality (RR 1.09, 95% CI 0.61 to 1.96, I² = 0%, 2 trials), myocardial infarction (RR 0.46, 95% CI 0.07 to 3.04, I² = 45%, 3 trials), unstable angina (RR 0.33, 95% CI 0.01 to 8.02, 1 trial), stroke (RR 1.03, 95% CI 0.07 to 16.25, 1 trial), adverse events (RR 0.89, 95% CI 0.76 to 1.05, I² = 0%, 2 trials), or any infection (rate ratio 0.66, 95% CI 0.32 to 1.36, I² = 0%, 4 trials). No trial assessed PVD or heart failure. TNF inhibitors The evidence is very uncertain about the effect of the intervention on all-cause mortality (RR 1.16, 95% CI 0.69 to 1.95, I² = 47%, 5 trials), heart failure (RR 0.92, 95% 0.75 to 1.14, I² = 0%, 4 trials), or adverse events (RR 1.15, 95% CI 0.84 to 1.56, I² = 32%, 2 trials). No trial assessed myocardial infarction, unstable angina, PVD or stroke. Adverse events may be underestimated and benefits inflated due to inadequate reporting. AUTHORS' CONCLUSIONS This Cochrane review assessed the benefits and harms of using interleukin-receptor antagonists and tumour necrosis factor inhibitors for primary and secondary prevention of atherosclerotic diseases compared with placebo or usual care. However, the evidence for the predetermined outcomes was deemed low or very low certainty, so there is still a need to determine whether these interventions provide clinical benefits or cause harm from this perspective. In summary, the different biases and imprecision in the included studies limit their external validity and represent a limitation to determining the effectiveness of the intervention for both primary and secondary prevention of ACVD.
Collapse
Key Words
- humans
- angina, unstable
- angina, unstable/mortality
- angina, unstable/prevention & control
- antibodies, monoclonal, humanized
- antibodies, monoclonal, humanized/administration & dosage
- antibodies, monoclonal, humanized/adverse effects
- atherosclerosis
- atherosclerosis/mortality
- atherosclerosis/prevention & control
- bias
- cause of death
- myocardial infarction
- myocardial infarction/mortality
- myocardial infarction/prevention & control
- primary prevention
- primary prevention/methods
- randomized controlled trials as topic
- receptors, interleukin-1
- receptors, interleukin-1/antagonists & inhibitors
- secondary prevention
- secondary prevention/methods
- tumor necrosis factor-alpha
- tumor necrosis factor-alpha/antagonists & inhibitors
Collapse
Affiliation(s)
- Arturo J Martí-Carvajal
- Universidad UTE, Facultad de Ciencias de la Salud Eugenio Espejo, Centro Asociado Cochrane Ecuador, Centro de Investigación en Salud Pública y Epidemiología Clínica (CISPEC), Quito, Ecuador
- Facultad de Medicina (Centro Cochrane Madrid), Universidad Francisco de Vitoria, Madrid, Spain
- Cátedra Rectoral de Medicina Basada en la Evidencia, Universidad de Carabobo, Valencia , Venezuela
| | - Mario A Gemmato-Valecillos
- Icahn School of Medicine at Mount Sinai/ NYCHH Elmhurst Hospital Center, 79-01 Broadway, Elmhurst, New York 11373, USA
| | | | - Mark Dayer
- Cardiovascular Research Institute, Mater Private Network, Dublin, Ireland
- Faculty of Health, University of Plymouth, Plymouth, UK
| | | | - Juan Bautista De Sanctis
- Institute of Molecular and Translational Medicine, Palacky University, Faculty of Medicine and Dentistry, Olomouc, Czech Republic
| | - Juan Marcos Parise Vasco
- Universidad UTE, Facultad de Ciencias de la Salud Eugenio Espejo, Centro Asociado Cochrane Ecuador, Centro de Investigación en Salud Pública y Epidemiología Clínica (CISPEC), Quito, Ecuador
| | - Ricardo J Riera Lizardo
- Cátedra Rectoral de Medicina Basada en la Evidencia, Universidad de Carabobo, Valencia, Venezuela
| | - Susana Nicola
- Universidad UTE, Facultad de Ciencias de la Salud Eugenio Espejo, Centro Asociado Cochrane Ecuador, Centro de Investigación en Salud Pública y Epidemiología Clínica (CISPEC), Quito, Ecuador
| | | | - Andrea Correa-Pérez
- Faculty of Medicine, Universidad Francisco de Vitoria, Madrid, Spain
- Hospital Pharmacy and Medical Devices Department, Hospital Central de la Defensa "Gómez Ulla" CSVE, Madrid, Spain
| |
Collapse
|
5
|
Lin Z, He J, Song C, Yuan S, Song Y, Bian X, Dou K. Association of triglyceride-glucose index with adverse cardiovascular events in patients with established coronary artery disease according to different inflammatory status. Nutr Metab Cardiovasc Dis 2024; 34:2124-2133. [PMID: 38749784 DOI: 10.1016/j.numecd.2024.04.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Revised: 04/18/2024] [Accepted: 04/19/2024] [Indexed: 08/06/2024]
Abstract
BACKGROUND AND AIMS This study aimed to investigate the association of the triglyceride-glucose (TyG) index, a simple-but-reliable indicator of insulin resistance, with risk of cardiovascular (CV) events in coronary artery disease (CAD) patients with different inflammation status. METHODS AND RESULTS We consecutively recruited 20,518 patients with angiograph-proven-CAD from 2017 to 2018 at Fuwai Hospital. Patients were categorized according to baseline TyG index tertiles (T) (tertile 1: ≤8.624; T2: 8.624-9.902 and T3: >9.902) and further assigned into 6 groups by high-sensitivity C-reactive protein (hsCRP) medians. The primary endpoint was CV events including CV death, nonfatal myocardial infarction and nonfatal stroke. During the 3.1-year-follow-up, 618 (3.0%) CV events were recorded. Overall, patients with high TyG index levels (T2 or T3) showed significantly increased risk of CV events (hazard ratio [HR]: 1.24; 95% confidence interval [CI]: 1.01-1.53; HR: 1.33; 95%CI: 1.05-1.68, respectively) compared with those with lowest Tyg index (T1) after adjusting for confounding factors. Upon stratification by hsCRP levels, elevated TyG index was associated with increased risk of CV events only in patients with hsCRP levels > median (per-1-unit-increase HR: 1.39; 95%CI: 1.11-1.74), rather than in those with hsCRP levels ≤ median. Furthermore, adding the TyG index to the predicting model led to a significant improvement in patients with hsCRP > median rather than in those with hsCRP ≤ median. CONCLUSIONS We firstly found that elevated TyG index levels were associated with increased risk of CV events in CAD patients, especially in those with increased inflammatory status, suggesting that it could help in risk stratification and prognosis in this population.
Collapse
Affiliation(s)
- Zhangyu Lin
- Cardiometabolic Medicine Center, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China; Department of Cardiology, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China; State Key Laboratory of Cardiovascular Disease, Beijing, China; National Clinical Research Center for Cardiovascular Diseases, Beijing, China
| | - Jining He
- Cardiometabolic Medicine Center, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China; Department of Cardiology, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China; State Key Laboratory of Cardiovascular Disease, Beijing, China; National Clinical Research Center for Cardiovascular Diseases, Beijing, China
| | - Chenxi Song
- Cardiometabolic Medicine Center, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China; Department of Cardiology, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China; State Key Laboratory of Cardiovascular Disease, Beijing, China; National Clinical Research Center for Cardiovascular Diseases, Beijing, China
| | - Sheng Yuan
- Cardiometabolic Medicine Center, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China; Department of Cardiology, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China; State Key Laboratory of Cardiovascular Disease, Beijing, China; National Clinical Research Center for Cardiovascular Diseases, Beijing, China
| | - Yanjun Song
- Cardiometabolic Medicine Center, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China; Department of Cardiology, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China; State Key Laboratory of Cardiovascular Disease, Beijing, China; National Clinical Research Center for Cardiovascular Diseases, Beijing, China
| | - Xiaohui Bian
- Cardiometabolic Medicine Center, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China; Department of Cardiology, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China; State Key Laboratory of Cardiovascular Disease, Beijing, China; National Clinical Research Center for Cardiovascular Diseases, Beijing, China
| | - Kefei Dou
- Cardiometabolic Medicine Center, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China; Department of Cardiology, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China; State Key Laboratory of Cardiovascular Disease, Beijing, China; National Clinical Research Center for Cardiovascular Diseases, Beijing, China.
| |
Collapse
|
6
|
Le R, Nguyen MT, Allahwala MA, Psaltis JP, Marathe CS, Marathe JA, Psaltis PJ. Cardiovascular Protective Properties of GLP-1 Receptor Agonists: More than Just Diabetic and Weight Loss Drugs. J Clin Med 2024; 13:4674. [PMID: 39200816 PMCID: PMC11355214 DOI: 10.3390/jcm13164674] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Revised: 08/06/2024] [Accepted: 08/07/2024] [Indexed: 09/02/2024] Open
Abstract
Owing to their potent glucose-lowering efficacy and substantial weight loss effects, glucagon-like peptide-1 receptor agonists (GLP-1 RAs) are now considered part of the frontline therapeutic options to treat both type 2 diabetes mellitus and nondiabetic overweight/obesity. Stemming from successful demonstration of their cardiometabolic modulation and reduction of major adverse cardiovascular events in clinical outcome trials, GLP-1 RAs have since been validated as agents with compelling cardiovascular protective properties. Studies spanning from the bench to preclinical and large-scale randomised controlled trials have consistently corroborated the cardiovascular benefits of this pharmacological class. Most notably, there is converging evidence that they exert favourable effects on atherosclerotic ischaemic endpoints, with preclinical data indicating that they may do so by directly modifying the burden and composition of atherosclerotic plaques. This narrative review examines the underlying pharmacology and clinical evidence behind the cardiovascular benefits of GLP-1 RAs, with particular focus on atherosclerotic cardiovascular disease. It also delves into the mechanisms that underpin their putative plaque-modifying actions, addresses existing knowledge gaps and therapeutic challenges and looks to future developments in the field, including the use of combination incretin agents for diabetes and weight loss management.
Collapse
Affiliation(s)
- Richard Le
- College of Medicine and Public Health, Flinders University, Adelaide 5042, Australia;
- Heart and Vascular Program, Lifelong Health Theme, South Australian Health and Medical Research Institute, Adelaide 5000, Australia; (M.T.N.); (M.A.A.); (J.A.M.)
| | - Mau T. Nguyen
- Heart and Vascular Program, Lifelong Health Theme, South Australian Health and Medical Research Institute, Adelaide 5000, Australia; (M.T.N.); (M.A.A.); (J.A.M.)
- Department of Cardiology, Central Adelaide Local Health Network, Adelaide 5000, Australia
- Adelaide Medical School, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide 5005, Australia; (J.P.P.); (C.S.M.)
| | - Momina A. Allahwala
- Heart and Vascular Program, Lifelong Health Theme, South Australian Health and Medical Research Institute, Adelaide 5000, Australia; (M.T.N.); (M.A.A.); (J.A.M.)
- Adelaide Medical School, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide 5005, Australia; (J.P.P.); (C.S.M.)
| | - James P. Psaltis
- Adelaide Medical School, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide 5005, Australia; (J.P.P.); (C.S.M.)
| | - Chinmay S. Marathe
- Adelaide Medical School, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide 5005, Australia; (J.P.P.); (C.S.M.)
- Department of Endocrinology, Central Adelaide Local Health Network, Adelaide 5000, Australia
| | - Jessica A. Marathe
- Heart and Vascular Program, Lifelong Health Theme, South Australian Health and Medical Research Institute, Adelaide 5000, Australia; (M.T.N.); (M.A.A.); (J.A.M.)
- Department of Cardiology, Central Adelaide Local Health Network, Adelaide 5000, Australia
- Adelaide Medical School, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide 5005, Australia; (J.P.P.); (C.S.M.)
| | - Peter J. Psaltis
- Heart and Vascular Program, Lifelong Health Theme, South Australian Health and Medical Research Institute, Adelaide 5000, Australia; (M.T.N.); (M.A.A.); (J.A.M.)
- Department of Cardiology, Central Adelaide Local Health Network, Adelaide 5000, Australia
- Adelaide Medical School, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide 5005, Australia; (J.P.P.); (C.S.M.)
| |
Collapse
|
7
|
Nagar N, Naidu G, Panda SK, Gulati K, Singh RP, Poluri KM. Elucidating the role of chemokines in inflammaging associated atherosclerotic cardiovascular diseases. Mech Ageing Dev 2024; 220:111944. [PMID: 38782074 DOI: 10.1016/j.mad.2024.111944] [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: 01/31/2024] [Revised: 05/08/2024] [Accepted: 05/14/2024] [Indexed: 05/25/2024]
Abstract
Age-related inflammation or inflammaging is a critical deciding factor of physiological homeostasis during aging. Cardiovascular diseases (CVDs) are exquisitely associated with aging and inflammation and are one of the leading causes of high mortality in the elderly population. Inflammaging comprises dysregulation of crosstalk between the vascular and cardiac tissues that deteriorates the vasculature network leading to development of atherosclerosis and atherosclerotic-associated CVDs in elderly populations. Leukocyte differentiation, migration and recruitment holds a crucial position in both inflammaging and atherosclerotic CVDs through relaying the activity of an intricate network of inflammation-associated protein-protein interactions. Among these interactions, small immunoproteins such as chemokines play a major role in the progression of inflammaging and atherosclerosis. Chemokines are actively involved in lymphocyte migration and severe inflammatory response at the site of injury. They relay their functions via chemokine-G protein-coupled receptors-glycosaminoglycan signaling axis and is a principal part for the detection of age-related atherosclerosis and related CVDs. This review focuses on highlighting the detailed intricacies of the effects of chemokine-receptor interaction and chemokine oligomerization on lymphocyte recruitment and its evident role in clinical manifestations of atherosclerosis and related CVDs. Further, the role of chemokine mediated signaling for formulating next-generation therapeutics against atherosclerosis has also been discussed.
Collapse
Affiliation(s)
- Nupur Nagar
- Department of Biosciences and Bioengineering, Indian Institute of Technology Roorkee, Roorkee, Uttarakhand 247667, India
| | - Goutami Naidu
- Department of Biosciences and Bioengineering, Indian Institute of Technology Roorkee, Roorkee, Uttarakhand 247667, India
| | - Santosh Kumar Panda
- Department of Biosciences and Bioengineering, Indian Institute of Technology Roorkee, Roorkee, Uttarakhand 247667, India
| | - Khushboo Gulati
- Department of Biosciences and Bioengineering, Indian Institute of Technology Roorkee, Roorkee, Uttarakhand 247667, India
| | - Ravindra Pal Singh
- Department of Industrial Biotechnology, Gujarat Biotechnology University, Gujarat International Finance Tec-City, Gandhinagar, Gujarat 382355, India
| | - Krishna Mohan Poluri
- Department of Biosciences and Bioengineering, Indian Institute of Technology Roorkee, Roorkee, Uttarakhand 247667, India; Centre for Nanotechnology, Indian Institute of Technology Roorkee, Roorkee, Uttarakhand 247667, India.
| |
Collapse
|
8
|
Stringham NT, Green M, Roche W, Prado-Cabrero A, Mulcahy R, Nolan J. Lutein, zeaxanthin, and meso-zeaxanthin supplementation attenuates inflammatory cytokines and markers of oxidative cardiovascular processes in humans. Nutr Metab Cardiovasc Dis 2024; 34:1976-1983. [PMID: 38890092 DOI: 10.1016/j.numecd.2024.05.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Revised: 05/03/2024] [Accepted: 05/06/2024] [Indexed: 06/20/2024]
Abstract
BACKGROUND AND AIMS Systemic inflammation and oxidation are primary contributors to the development of atherosclerosis. Oxidation of low-density lipoprotein (LDL) particles within the vascular endothelium has been hypothesized to be an initial step in the formation of atherosclerotic plaques, with inflammatory cytokines serving as the signaling mechanism for concomitant macrophage activation. Supplementation with the antioxidative macular xanthophylls (lutein [L], zeaxanthin [Z], and meso-zeaxanthin [MZ]) has been shown to aid in the reduction of inflammatory physiologic responses; therefore, we hypothesized that in our study population, supplementation with these xanthophylls would facilitate a systemic reduction in markers of inflammation and cardiovascular lipid oxidation. METHODS AND RESULTS In this double-blind placebo-controlled supplementation study, participants were randomly allocated to receive the active intervention containing L (10 mg) + MZ (10 mg) + Z (2 mg) or placebo (containing sunflower oil). Serum concentrations of carotenoids (assessed by HPLC), inflammatory cytokines (IL-6, IL-1β, TNF-α) and oxidized LDL (OxLDL; by solid-phase sandwich ELISA) were measured at baseline and at 6-months. Results showed that over the supplementation period, compared to placebo, the active group demonstrated statistically significant increases in serum concentrations of L, Z, & MZ (p < 0.05), reductions in inflammatory cytokines IL-1β (p < 0.001) and TNF-α (p = 0.003), as well as a corresponding reduction in serum OxLDL (p = 0.009). CONCLUSIONS Our data show that L, Z, & MZ supplementation results in decreased serum IL-1β, TNF-α, and OxLDL. This suggests that these carotenoids are acting systemically to attenuate oxidative lipid products and inflammation, thus reducing their contribution to atherosclerotic plaque formation.
Collapse
Affiliation(s)
- Nicole T Stringham
- Nutrition Research Centre Ireland (NRCI), Southeast Technical University, Waterford, Ireland; Northern Arizona University, Flagstaff, AZ, USA.
| | - Marina Green
- Nutrition Research Centre Ireland (NRCI), Southeast Technical University, Waterford, Ireland
| | - Warren Roche
- Nutrition Research Centre Ireland (NRCI), Southeast Technical University, Waterford, Ireland
| | - Alfonso Prado-Cabrero
- Nutrition Research Centre Ireland (NRCI), Southeast Technical University, Waterford, Ireland
| | - Riona Mulcahy
- Nutrition Research Centre Ireland (NRCI), Southeast Technical University, Waterford, Ireland
| | - John Nolan
- Nutrition Research Centre Ireland (NRCI), Southeast Technical University, Waterford, Ireland
| |
Collapse
|
9
|
Gheysari R, Nikbaf-Shandiz M, Hosseini AM, Rasaei N, Hosseini S, Bahari H, Asbaghi O, Rastgoo S, Goudarzi K, Shiraseb F, Behmadi R. The effects of L-carnitine supplementation on cardiovascular risk factors in participants with impaired glucose tolerance and diabetes: a systematic review and dose-response meta-analysis. Diabetol Metab Syndr 2024; 16:185. [PMID: 39085907 PMCID: PMC11290177 DOI: 10.1186/s13098-024-01415-8] [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: 01/11/2024] [Accepted: 07/16/2024] [Indexed: 08/02/2024] Open
Abstract
AIMS L-carnitine plays a role related to cardiometabolic factors, but its effectiveness and safety in CVD are still unknown. We aim to assess the effect of L-carnitine supplementation on CVD risk factors. METHODS A systematic literature search was conducted in PubMed, Web of Science, and Scopus until October 2022. The main outcomes were lipid profiles, anthropometric parameters, insulin resistance, serum glucose levels, leptin, blood pressure, and inflammatory markers. The pooled weighted mean difference (WMD) was calculated using a random-effects model. RESULTS We included the 21 RCTs (n = 2900) with 21 effect sizes in this study. L-carnitine supplementation had a significant effect on TG (WMD = - 13.50 mg/dl, p = 0.039), LDL (WMD = - 12.66 mg/dl, p < 0.001), FBG (WMD = - 6.24 mg/dl, p = 0.001), HbA1c (WMD = -0.37%, p = 0.013) HOMA-IR (WMD = -0.72, p = 0.038 (, CRP (WMD = - 0.07 mg/dl, P = 0.037), TNF-α (WMD = - 1.39 pg/ml, p = 0.033), weight (WMD = - 1.58 kg, p = 0.001 (, BMI (WMD = - 0.28 kg/m2, p = 0.017(, BFP (WMD = - 1.83, p < 0.001) and leptin (WMD = - 2.21 ng/ml, p = 0.003 (in intervention, compared to the placebo group, in the pooled analysis. CONCLUSIONS This meta-analysis demonstrated that administration of L-carnitine in diabetic and glucose intolerance patients can significantly reduce TG, LDL-C, FBG, HbA1c, HOMA-IR, CRP, TNF-α, weight, BMI, BFP, and leptin levels. PROSPERO registration code: CRD42022366992.
Collapse
Affiliation(s)
- Rezvan Gheysari
- Shohada-E-Tajrish Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | | | - Amir Mehdi Hosseini
- Faculty of Medical Sciences and Technologies, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Niloufar Rasaei
- Department of Community Nutrition, School of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences (TUMS), Tehran, Iran
| | - Shabnam Hosseini
- School of Human Nutrition, Faculty of Agricultural and Environmental Sciences, McGill University, Montreal, QC, Canada
| | - Hossein Bahari
- Transplant Research Center, Clinical Research Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Omid Asbaghi
- Cancer Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Student Research Committee, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Samira Rastgoo
- Student Research Committee, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Department of Cellular and Molecular Nutrition, Faculty of Nutrition Science and Food Technology, National Nutrition and Food Technology Research Institute, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Kian Goudarzi
- Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Farideh Shiraseb
- Department of Community Nutrition, School of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences (TUMS), Tehran, Iran.
| | - Reza Behmadi
- Department of Pediatrics, School of Medicine, Iran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
10
|
Fan Q, Tan Z, Su W, Li Q, Jin D, Du Y, Zhang L, Wu S. Efficacy, safety and mechanism of Simiaoyongan decoction in the treatment of carotid atherosclerotic plaque: a randomized, double-blind, placebo-controlled clinical trial protocol. BMC Complement Med Ther 2024; 24:277. [PMID: 39039498 PMCID: PMC11265120 DOI: 10.1186/s12906-024-04555-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2023] [Accepted: 06/17/2024] [Indexed: 07/24/2024] Open
Abstract
INTRODUCTION Chronic inflammation is the major pathological feature of Atherosclerosis(As). Inflammation may accelerate plaque to develop, which is a key factor resulting in the thinning of the fibrous cap and the vulnerable rupture of plaque. Presently, clinical treatments are still lacking. It is necessary to find a safe and effective treatment for As inflammation. Simiaoyongan Decoction (SMYA) has potential anti-inflammatory and plaque protection effects. This protocol aims to evaluate the efficacy, safety, and mechanism of SMYA for patients with carotid atherosclerotic plaque. METHODS/DESIGN The assessment of SMYA clinical trial is designed as a randomized, double-blind, placebo-controlled study. The sample size is 86 cases in total, with 43 participants in the intervention group and the control group respectively. The intervention group takes SMYA, while the control group takes SMYA placebo. The medication lasts for 14 days every 10 weeks, with a total of 50 weeks. We will use carotid artery high resolution magnetic resonance imaging (HR-MRI) to measure plaque. The plaque minimum fiber cap thickness (PMFCT) is adopted as the primary outcome. The secondary outcomes include plaque fiber cap volume, volume percentage of fiber cap, lipid-rich necrotic core (LRNC) volume, volume percentage of LRNC, internal bleeding volume of plaque, internal bleeding volume percentage of plaque, plaque calcification volume, volume percentage of plaque calcification, lumen stenosis rate, average and a maximum of vessel wall thickness, vessel wall volume, total vessel wall load, carotid atherosclerosis score, hs-CRP, IL-1β and IL-6, the level of lipid profiles and blood glucose, blood pressure, and body weight. DISCUSSION We anticipate that patients with As plaque will be improved from SMYA by inhibiting inflammation to enhance plaque stability. This study analyzes plaque by using HR-MRI to evaluate the clinical efficacy and safety of SMYA. Moreover, we conduct transcriptome analysis, proteomic analysis, and metagenomic analysis of blood and stool of participants to study the mechanism of SMYA against As plaque. This is the first prospective TCM trial to observe and treat As plaque by inhibiting inflammatory reaction directly. If successful, the finding will be valuable in the treatment of As plaque and drug development, especially in the "statin era". TRIAL REGISTRATION NUMBER This trial is registered on Chinese Clinical Trials.gov with number ChiCTR2000039062 on October 15, 2020 ( http://www.chictr.org.cn ).
Collapse
Affiliation(s)
- QinHua Fan
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - ZhongJian Tan
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - WenQuan Su
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - QingXiao Li
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Dian Jin
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - YaWei Du
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - LiPing Zhang
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China.
| | - ShengXian Wu
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China.
| |
Collapse
|
11
|
Nankivell V, Vidanapathirana AK, Hoogendoorn A, Tan JTM, Verjans J, Psaltis PJ, Hutchinson MR, Gibson BC, Lu Y, Goldys E, Zheng G, Bursill CA. Targeting macrophages with multifunctional nanoparticles to detect and prevent atherosclerotic cardiovascular disease. Cardiovasc Res 2024; 120:819-838. [PMID: 38696700 PMCID: PMC11218693 DOI: 10.1093/cvr/cvae099] [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: 09/28/2023] [Revised: 02/29/2024] [Accepted: 04/02/2024] [Indexed: 05/04/2024] Open
Abstract
Despite the emergence of novel diagnostic, pharmacological, interventional, and prevention strategies, atherosclerotic cardiovascular disease remains a significant cause of morbidity and mortality. Nanoparticle (NP)-based platforms encompass diverse imaging, delivery, and pharmacological properties that provide novel opportunities for refining diagnostic and therapeutic interventions for atherosclerosis at the cellular and molecular levels. Macrophages play a critical role in atherosclerosis and therefore represent an important disease-related diagnostic and therapeutic target, especially given their inherent ability for passive and active NP uptake. In this review, we discuss an array of inorganic, carbon-based, and lipid-based NPs that provide magnetic, radiographic, and fluorescent imaging capabilities for a range of highly promising research and clinical applications in atherosclerosis. We discuss the design of NPs that target a range of macrophage-related functions such as lipoprotein oxidation, cholesterol efflux, vascular inflammation, and defective efferocytosis. We also provide examples of NP systems that were developed for other pathologies such as cancer and highlight their potential for repurposing in cardiovascular disease. Finally, we discuss the current state of play and the future of theranostic NPs. Whilst this is not without its challenges, the array of multifunctional capabilities that are possible in NP design ensures they will be part of the next frontier of exciting new therapies that simultaneously improve the accuracy of plaque diagnosis and more effectively reduce atherosclerosis with limited side effects.
Collapse
Affiliation(s)
- Victoria Nankivell
- Australian Research Council (ARC) Centre of Excellence for Nanoscale BioPhotonics (CNBP)
- Vascular Research Centre, Lifelong Health, South Australian Health and Medical Research Institute (SAHMRI), North Terrace, Adelaide, 5000, Australia
- Faculty of Health and Medical Science, The University of Adelaide, North Terrace, Adelaide, 5000, Australia
| | - Achini K Vidanapathirana
- Australian Research Council (ARC) Centre of Excellence for Nanoscale BioPhotonics (CNBP)
- Vascular Research Centre, Lifelong Health, South Australian Health and Medical Research Institute (SAHMRI), North Terrace, Adelaide, 5000, Australia
- Faculty of Health and Medical Science, The University of Adelaide, North Terrace, Adelaide, 5000, Australia
| | - Ayla Hoogendoorn
- Australian Research Council (ARC) Centre of Excellence for Nanoscale BioPhotonics (CNBP)
- Vascular Research Centre, Lifelong Health, South Australian Health and Medical Research Institute (SAHMRI), North Terrace, Adelaide, 5000, Australia
| | - Joanne T M Tan
- Vascular Research Centre, Lifelong Health, South Australian Health and Medical Research Institute (SAHMRI), North Terrace, Adelaide, 5000, Australia
- Faculty of Health and Medical Science, The University of Adelaide, North Terrace, Adelaide, 5000, Australia
| | - Johan Verjans
- Australian Research Council (ARC) Centre of Excellence for Nanoscale BioPhotonics (CNBP)
- Vascular Research Centre, Lifelong Health, South Australian Health and Medical Research Institute (SAHMRI), North Terrace, Adelaide, 5000, Australia
- Faculty of Health and Medical Science, The University of Adelaide, North Terrace, Adelaide, 5000, Australia
| | - Peter J Psaltis
- Australian Research Council (ARC) Centre of Excellence for Nanoscale BioPhotonics (CNBP)
- Vascular Research Centre, Lifelong Health, South Australian Health and Medical Research Institute (SAHMRI), North Terrace, Adelaide, 5000, Australia
- Faculty of Health and Medical Science, The University of Adelaide, North Terrace, Adelaide, 5000, Australia
| | - Mark R Hutchinson
- Australian Research Council (ARC) Centre of Excellence for Nanoscale BioPhotonics (CNBP)
- Faculty of Health and Medical Science, The University of Adelaide, North Terrace, Adelaide, 5000, Australia
| | - Brant C Gibson
- Australian Research Council (ARC) Centre of Excellence for Nanoscale BioPhotonics (CNBP)
- School of Science, RMIT University, Melbourne, Victoria, Australia
| | - Yiqing Lu
- Australian Research Council (ARC) Centre of Excellence for Nanoscale BioPhotonics (CNBP)
- School of Engineering, Macquarie University, Sydney, NSW, Australia
| | - Ewa Goldys
- Australian Research Council (ARC) Centre of Excellence for Nanoscale BioPhotonics (CNBP)
- Graduate School of Biomedical Engineering, University of New South Wales, High Street, NSW, 2052, Australia
| | - Gang Zheng
- Australian Research Council (ARC) Centre of Excellence for Nanoscale BioPhotonics (CNBP)
- Department of Medical Biophysics, University of Toronto, 101 College Street, Toronto, M5G 1L7, Canada
| | - Christina A Bursill
- Australian Research Council (ARC) Centre of Excellence for Nanoscale BioPhotonics (CNBP)
- Vascular Research Centre, Lifelong Health, South Australian Health and Medical Research Institute (SAHMRI), North Terrace, Adelaide, 5000, Australia
- Faculty of Health and Medical Science, The University of Adelaide, North Terrace, Adelaide, 5000, Australia
| |
Collapse
|
12
|
Karnewar S, Karnewar V, Deaton R, Shankman LS, Benavente ED, Williams CM, Bradley X, Alencar GF, Bulut GB, Kirmani S, Baylis RA, Zunder ER, den Ruijter HM, Pasterkamp G, Owens GK. IL-1β Inhibition Partially Negates the Beneficial Effects of Diet-Induced Atherosclerosis Regression in Mice. Arterioscler Thromb Vasc Biol 2024; 44:1379-1392. [PMID: 38695167 PMCID: PMC11111338 DOI: 10.1161/atvbaha.124.320800] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Accepted: 04/16/2024] [Indexed: 05/14/2024]
Abstract
BACKGROUND Thromboembolic events secondary to rupture or erosion of advanced atherosclerotic lesions is the global leading cause of death. The most common and effective means to reduce these major adverse cardiovascular events, including myocardial infarction and stroke, is aggressive lipid lowering via a combination of drugs and dietary modifications. However, we know little regarding the effects of reducing dietary lipids on the composition and stability of advanced atherosclerotic lesions, the mechanisms that regulate these processes, and what therapeutic approaches might augment the benefits of lipid lowering. METHODS Smooth muscle cell lineage-tracing Apoe-/- mice were fed a high-cholesterol Western diet for 18 weeks and then a zero-cholesterol standard laboratory diet for 12 weeks before treating them with an IL (interleukin)-1β or control antibody for 8 weeks. We assessed lesion size and remodeling indices, as well as the cellular composition of aortic and brachiocephalic artery lesions, indices of plaque stability, overall plaque burden, and phenotypic transitions of smooth muscle cell and other lesion cells by smooth muscle cell lineage tracing combined with single-cell RNA sequencing, cytometry by time-of-flight, and immunostaining plus high-resolution confocal microscopic z-stack analysis. RESULTS Lipid lowering by switching Apoe-/- mice from a Western diet to a standard laboratory diet reduced LDL cholesterol levels by 70% and resulted in multiple beneficial effects including reduced overall aortic plaque burden, as well as reduced intraplaque hemorrhage and necrotic core area. However, contrary to expectations, IL-1β antibody treatment after diet-induced reductions in lipids resulted in multiple detrimental changes including increased plaque burden and brachiocephalic artery lesion size, as well as increasedintraplaque hemorrhage, necrotic core area, and senescence as compared with IgG control antibody-treated mice. Furthermore, IL-1β antibody treatment upregulated neutrophil degranulation pathways but downregulated smooth muscle cell extracellular matrix pathways likely important for the protective fibrous cap. CONCLUSIONS Taken together, IL-1β appears to be required for the maintenance of standard laboratory diet-induced reductions in plaque burden and increases in multiple indices of plaque stability.
Collapse
MESH Headings
- Animals
- Interleukin-1beta/metabolism
- Atherosclerosis/pathology
- Atherosclerosis/prevention & control
- Atherosclerosis/metabolism
- Atherosclerosis/genetics
- Disease Models, Animal
- Plaque, Atherosclerotic
- Mice
- Myocytes, Smooth Muscle/pathology
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/drug effects
- Mice, Knockout, ApoE
- Male
- Diet, Western
- Mice, Inbred C57BL
- Aorta/pathology
- Aorta/metabolism
- Aorta/drug effects
- Aortic Diseases/pathology
- Aortic Diseases/prevention & control
- Aortic Diseases/genetics
- Aortic Diseases/metabolism
- Diet, High-Fat
- Muscle, Smooth, Vascular/pathology
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/drug effects
- Brachiocephalic Trunk/pathology
- Brachiocephalic Trunk/metabolism
- Brachiocephalic Trunk/drug effects
Collapse
Affiliation(s)
- Santosh Karnewar
- Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville, USA
| | - Vaishnavi Karnewar
- Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville, USA
| | - Rebecca Deaton
- Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville, USA
| | - Laura S. Shankman
- Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville, USA
| | - Ernest D. Benavente
- Laboratory of Experimental Cardiology, University Medical Center Utrecht, Utrecht University, the Netherlands
| | - Corey M. Williams
- Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville, USA
| | - Xenia Bradley
- Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville, USA
| | - Gabriel F. Alencar
- Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville, USA
| | - Gamze B. Bulut
- Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville, USA
| | - Sara Kirmani
- Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville, USA
| | - Richard A. Baylis
- Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville, USA
| | - Eli R. Zunder
- Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville, USA
| | - Hester M. den Ruijter
- Laboratory of Experimental Cardiology, University Medical Center Utrecht, Utrecht University, the Netherlands
| | - Gerard Pasterkamp
- Laboratory of Experimental Cardiology, University Medical Center Utrecht, Utrecht University, the Netherlands
| | - Gary K. Owens
- Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville, USA
| |
Collapse
|
13
|
Liao M, Liu L, Bai L, Wang R, Liu Y, Zhang L, Han J, Li Y, Qi B. Correlation between novel inflammatory markers and carotid atherosclerosis: A retrospective case-control study. PLoS One 2024; 19:e0303869. [PMID: 38809853 PMCID: PMC11135694 DOI: 10.1371/journal.pone.0303869] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Accepted: 05/01/2024] [Indexed: 05/31/2024] Open
Abstract
OBJECTIVE Carotid atherosclerosis is a chronic inflammatory disease, which is a major cause of ischemic stroke. The purpose of this study was to analyze the relationship between carotid atherosclerosis and novel inflammatory markers, including platelet to lymphocyte ratio (PLR), neutrophil to lymphocyte ratio (NLR), lymphocyte to monocyte ratio (LMR), platelet to neutrophil ratio (PNR), neutrophil to lymphocyte platelet ratio (NLPR), systemic immune-inflammation index (SII), systemic inflammation response index (SIRI), and aggregate index of systemic inflammation (AISI), in order to find the best inflammatory predictor of carotid atherosclerosis. METHOD We included 10015 patients who underwent routine physical examinations at the physical examination center of our hospital from January 2016 to December 2019, among whom 1910 were diagnosed with carotid atherosclerosis. The relationship between novel inflammatory markers and carotid atherosclerosis was analyzed by logistic regression, and the effectiveness of each factor in predicting carotid atherosclerosis was evaluated by receiver operating characteristic (ROC) curve and area under the curve (AUC). RESULT The level of PLR, LMR and PNR in the carotid atherosclerosis group were lower than those in the non-carotid atherosclerosis group, while NLR, NLPR, SII, SIRI and AISI in the carotid atherosclerosis group were significantly higher than those in the non-carotid atherosclerosis group. Logistic regression analysis showed that PLR, NLR, LMR, PNR, NLPR, SII, SIRI, AISI were all correlated with carotid atherosclerosis. The AUC value of NLPR was the highest, which was 0.67, the cut-off value was 0.78, the sensitivity was 65.8%, and the specificity was 57.3%. The prevalence rate of carotid atherosclerosis was 12.4% below the cut-off, 26.6% higher than the cut-off, and the prevalence rate increased by 114.5%. CONCLUSION New inflammatory markers were significantly correlated with carotid atherosclerosis, among which NLPR was the optimum inflammatory marker to predict the risk of carotid atherosclerosis.
Collapse
Affiliation(s)
- Man Liao
- Department of General, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Lihua Liu
- Department of General, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Lijuan Bai
- Department of General, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Ruiyun Wang
- Department of General, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Yun Liu
- Department of General, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Liting Zhang
- Department of General, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Jing Han
- Department of General, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Yunqiao Li
- Department of General, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Benling Qi
- Department of General, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| |
Collapse
|
14
|
Lin Y, Xie R, Yu T. Photodynamic Therapy for Atherosclerosis: Past, Present, and Future. Pharmaceutics 2024; 16:729. [PMID: 38931851 PMCID: PMC11206729 DOI: 10.3390/pharmaceutics16060729] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2024] [Revised: 05/22/2024] [Accepted: 05/24/2024] [Indexed: 06/28/2024] Open
Abstract
This review paper examines the evolution of photodynamic therapy (PDT) as a novel, minimally invasive strategy for treating atherosclerosis, a leading global health concern. Atherosclerosis is characterized by the accumulation of lipids and inflammation within arterial walls, leading to significant morbidity and mortality through cardiovascular diseases such as myocardial infarction and stroke. Traditional therapeutic approaches have primarily focused on modulating risk factors such as hypertension and hyperlipidemia, with emerging evidence highlighting the pivotal role of inflammation. PDT, leveraging a photosensitizer, specific-wavelength light, and oxygen, offers targeted treatment by inducing cell death in diseased tissues while sparing healthy ones. This specificity, combined with advancements in nanoparticle technology for improved delivery, positions PDT as a promising alternative to traditional interventions. The review explores the mechanistic basis of PDT, its efficacy in preclinical studies, and the potential for enhancing plaque stability and reducing macrophage density within plaques. It also addresses the need for further research to optimize treatment parameters, mitigate adverse effects, and validate long-term outcomes. By detailing past developments, current progress, and future directions, this paper aims to highlight PDT's potential in revolutionizing atherosclerosis treatment, bridging the gap from experimental research to clinical application.
Collapse
Affiliation(s)
- Yanqing Lin
- Ultrasound in Cardiac Electrophysiology and Biomechanics Key Laboratory of Sichuan Province, Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu 610072, China;
| | - Ruosen Xie
- Wisconsin Institute for Discovery, University of Wisconsin–Madison, Madison, WI 53705, USA;
| | - Tao Yu
- Department of Cardiac Surgery, Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu 610072, China
| |
Collapse
|
15
|
Fularski P, Czarnik W, Dąbek B, Lisińska W, Radzioch E, Witkowska A, Młynarska E, Rysz J, Franczyk B. Broader Perspective on Atherosclerosis-Selected Risk Factors, Biomarkers, and Therapeutic Approach. Int J Mol Sci 2024; 25:5212. [PMID: 38791250 PMCID: PMC11121693 DOI: 10.3390/ijms25105212] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2024] [Revised: 05/01/2024] [Accepted: 05/07/2024] [Indexed: 05/26/2024] Open
Abstract
Atherosclerotic cardiovascular disease (ASCVD) stands as the leading cause of mortality worldwide. At its core lies a progressive process of atherosclerosis, influenced by multiple factors. Among them, lifestyle-related factors are highlighted, with inadequate diet being one of the foremost, alongside factors such as cigarette smoking, low physical activity, and sleep deprivation. Another substantial group of risk factors comprises comorbidities. Amongst others, conditions such as hypertension, diabetes mellitus (DM), chronic kidney disease (CKD), or familial hypercholesterolemia (FH) are included here. Extremely significant in the context of halting progression is counteracting the mentioned risk factors, including through treatment of the underlying disease. What is more, in recent years, there has been increasing attention paid to perceiving atherosclerosis as an inflammation-related disease. Consequently, efforts are directed towards exploring new anti-inflammatory medications to limit ASCVD progression. Simultaneously, research is underway to identify biomarkers capable of providing insights into the ongoing process of atherosclerotic plaque formation. The aim of this study is to provide a broader perspective on ASCVD, particularly focusing on its characteristics, traditional and novel treatment methods, and biomarkers that can facilitate its early detection.
Collapse
Affiliation(s)
- Piotr Fularski
- Department of Nephrocardiology, Medical University of Lodz, ul. Zeromskiego 113, 90-549 Lodz, Poland
| | - Witold Czarnik
- Department of Nephrocardiology, Medical University of Lodz, ul. Zeromskiego 113, 90-549 Lodz, Poland
| | - Bartłomiej Dąbek
- Department of Nephrocardiology, Medical University of Lodz, ul. Zeromskiego 113, 90-549 Lodz, Poland
| | - Wiktoria Lisińska
- Department of Nephrocardiology, Medical University of Lodz, ul. Zeromskiego 113, 90-549 Lodz, Poland
| | - Ewa Radzioch
- Department of Nephrocardiology, Medical University of Lodz, ul. Zeromskiego 113, 90-549 Lodz, Poland
| | - Alicja Witkowska
- Department of Nephrocardiology, Medical University of Lodz, ul. Zeromskiego 113, 90-549 Lodz, Poland
| | - Ewelina Młynarska
- Department of Nephrocardiology, Medical University of Lodz, ul. Zeromskiego 113, 90-549 Lodz, Poland
| | - Jacek Rysz
- Department of Nephrology, Hypertension and Family Medicine, Medical University of Lodz, ul. Zeromskiego 113, 90-549 Lodz, Poland
| | - Beata Franczyk
- Department of Nephrocardiology, Medical University of Lodz, ul. Zeromskiego 113, 90-549 Lodz, Poland
| |
Collapse
|
16
|
Liu Y, Dai L, Dong Y, Ma C, Cheng P, Jiang C, Liao H, Li Y, Wang X, Xu X. Coronary inflammation based on pericoronary adipose tissue attenuation in type 2 diabetic mellitus: effect of diabetes management. Cardiovasc Diabetol 2024; 23:108. [PMID: 38553738 PMCID: PMC10981289 DOI: 10.1186/s12933-024-02199-x] [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: 12/20/2023] [Accepted: 03/12/2024] [Indexed: 04/01/2024] Open
Abstract
BACKGROUND Coronary inflammation plays crucial role in type 2 diabetes mellitus (T2DM) induced cardiovascular complications. Both glucose-lowering drug interventions (GLDIS) and glycemic control (GC) status potentially correlate coronary inflammation, as indicated by changes in pericoronary adipose tissue (PCAT) attenuation, and thus influence cardiovascular risk. This study evaluated the impact of GLDIS and GC status on PCAT attenuation in T2DM patients. METHODS This retrospective study collected clinical data and coronary computed tomography angiography (CCTA) images of 1,342 patients, including 547 T2DM patients and 795 non-T2DM patients in two tertiary hospitals. T2DM patients were subgroup based on two criteria: (1) GC status: well: HbA1c < 7%, moderate: 7 ≤ HbA1c ≤ 9%, and poor: HbA1c > 9%; (2) GLDIS and non-GLDIS. PCAT attenuations of the left anterior descending artery (LAD-PCAT), left circumflex artery (LCX-PCAT), and right coronary artery (RCA-PCAT) were measured. Propensity matching (PSM) was used to cross compare PCAT attenuation of non-T2DM and all subgroups of T2DM patients. Linear regressions were conducted to evaluate the impact of GC status and GLDIS on PCAT attenuation in T2DM patients. RESULTS Significant differences were observed in RCA-PCAT and LCX-PCAT between poor GC-T2DM and non-T2DM patients (LCX: - 68.75 ± 7.59 HU vs. - 71.93 ± 7.25 HU, p = 0.008; RCA: - 74.37 ± 8.44 HU vs. - 77.2 ± 7.42 HU, p = 0.026). Higher PCAT attenuation was observed in LAD-PCAT, LCX-PCAT, and RCA-PCAT in non-GLDIS T2DM patients compared with GLDIS T2DM patients (LAD: - 78.11 ± 8.01 HU vs. - 75.04 ± 8.26 HU, p = 0.022; LCX: - 71.10 ± 8.13 HU vs. - 68.31 ± 7.90 HU, p = 0.037; RCA: - 78.17 ± 8.64 HU vs. - 73.35 ± 9.32 HU, p = 0.001). In the linear regression, other than sex and duration of diabetes, both metformin and acarbose were found to be significantly associated with lower LAD-PCAT (metformin: β coefficient = - 2.476, p=0.021; acarbose: β coefficient = - 1.841, p = 0.031). CONCLUSION Inadequate diabetes management, including poor GC and lack of GLDIS, may be associated with increased coronary artery inflammation in T2DM patients, as indicated by PCAT attenuation on CCTA, leading to increased cardiovascular risk. This finding could help healthcare providers identify T2DM patients with increased cardiovascular risk, develop improved cardiovascular management programs, and reduce subsequent cardiovascular related mortality.
Collapse
Affiliation(s)
- Yuankang Liu
- Department of Radiology, Liyuan Hospital, Tongji Medical College, Huazhong University of Science and Technology, 39 Yanhu Avenue, Wuchang District, Wuhan, 430077, China
| | - Lisong Dai
- Department of Diagnostic and Interventional Radiology, Shanghai Sixth People's Hospital, Shanghai Jiao Tong University School of Medicine, No. 600, Yishan Road, Xuhui District, Shanghai, Shanghai, 200233, China
| | - Yue Dong
- Department of Radiology, Liyuan Hospital, Tongji Medical College, Huazhong University of Science and Technology, 39 Yanhu Avenue, Wuchang District, Wuhan, 430077, China
| | - Cong Ma
- Department of Surgery, Liyuan Hospital, Tongji Medical College, Huazhong University of Science and Technology, 39 Yanhu Avenue, Wuchang District, Wuhan, 430077, China
| | - Panpan Cheng
- Department of Radiology, Liyuan Hospital, Tongji Medical College, Huazhong University of Science and Technology, 39 Yanhu Avenue, Wuchang District, Wuhan, 430077, China
| | - Cuiping Jiang
- Department of Radiology, Liyuan Hospital, Tongji Medical College, Huazhong University of Science and Technology, 39 Yanhu Avenue, Wuchang District, Wuhan, 430077, China
| | - Hongli Liao
- Department of Radiology, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Shengli Road No. 26, Jiangan District, Wuhan, 430014, China
| | - Ying Li
- Department of Radiology, Liyuan Hospital, Tongji Medical College, Huazhong University of Science and Technology, 39 Yanhu Avenue, Wuchang District, Wuhan, 430077, China
| | - Xiang Wang
- Department of Radiology, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Shengli Road No. 26, Jiangan District, Wuhan, 430014, China.
| | - Xiangyang Xu
- Department of Radiology, Liyuan Hospital, Tongji Medical College, Huazhong University of Science and Technology, 39 Yanhu Avenue, Wuchang District, Wuhan, 430077, China.
| |
Collapse
|
17
|
Attiq A, Afzal S, Ahmad W, Kandeel M. Hegemony of inflammation in atherosclerosis and coronary artery disease. Eur J Pharmacol 2024; 966:176338. [PMID: 38242225 DOI: 10.1016/j.ejphar.2024.176338] [Citation(s) in RCA: 31] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Revised: 12/30/2023] [Accepted: 01/16/2024] [Indexed: 01/21/2024]
Abstract
Inflammation drives coronary artery disease and atherosclerosis implications. Lipoprotein entry, retention, and oxidative modification cause endothelial damage, triggering innate and adaptive immune responses. Recruited immune cells orchestrate the early atherosclerotic lesions by releasing proinflammatory cytokines, expediting the foam cell formation, intraplaque haemorrhage, secretion of matrix-degrading enzymes, and lesion progression, eventually promoting coronary artery syndrome via various inflammatory cascades. In addition, soluble mediators disrupt the dynamic anti- and prothrombotic balance maintained by endothelial cells and pave the way for coronary artery disease such as angina pectoris. Recent studies have established a relationship between elevated levels of inflammatory markers, including C-reactive protein (CRP), interleukins (IL-6, IL-1β), and tumour necrosis factor-alpha (TNF-α) with the severity of CAD and the possibility of future cardiovascular events. High-sensitivity C-reactive protein (hs-CRP) is a marker for assessing systemic inflammation and predicting the risk of developing CAD based on its peak plasma levels. Hence, understanding cross-talk interactions of inflammation, atherogenesis, and CAD is highly warranted to recalculate the risk factors that activate and propagate arterial lesions and devise therapeutic strategies accordingly. Cholesterol-inflammation lowering agents (statins), monoclonal antibodies targeting IL-1 and IL-6 (canakinumab and tocilizumab), disease-modifying antirheumatic drugs (methotrexate), sodium-glucose transport protein-2 (SGLT2) inhibitors, colchicine and xanthene oxidase inhibitor (allopurinol) have shown promising results in reducing inflammation, regressing atherogenic plaque and modifying the course of CAD. Here, we review the complex interplay between inflammatory, endothelial, smooth muscle and foam cells. Moreover, the putative role of inflammation in atherosclerotic CAD, underlying mechanisms and potential therapeutic implications are also discussed herein.
Collapse
Affiliation(s)
- Ali Attiq
- Discipline of Pharmacology, School of Pharmaceutical Sciences, Universiti Sains Malaysia, Gelugor, 11800, Penang, Malaysia.
| | - Sheryar Afzal
- Department of Biomedical Sciences, College of Veterinary Medicine, King Faisal University, 31982, Al Ahsa, Saudi Arabia.
| | - Waqas Ahmad
- Discipline of Pharmaceutical Chemistry, School of Pharmaceutical Sciences, Universiti Sains Malaysia, Gelugor, 11800, Penang, Malaysia
| | - Mahmoud Kandeel
- Department of Biomedical Sciences, College of Veterinary Medicine, King Faisal University, 31982, Al Ahsa, Saudi Arabia
| |
Collapse
|
18
|
Kounatidis D, Vallianou NG, Poulaki A, Evangelopoulos A, Panagopoulos F, Stratigou T, Geladari E, Karampela I, Dalamaga M. ApoB100 and Atherosclerosis: What's New in the 21st Century? Metabolites 2024; 14:123. [PMID: 38393015 PMCID: PMC10890411 DOI: 10.3390/metabo14020123] [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: 01/04/2024] [Revised: 02/08/2024] [Accepted: 02/09/2024] [Indexed: 02/25/2024] Open
Abstract
ApoB is the main protein of triglyceride-rich lipoproteins and is further divided into ApoB48 in the intestine and ApoB100 in the liver. Very low-density lipoprotein (VLDL) is produced by the liver, contains ApoB100, and is metabolized into its remnants, intermediate-density lipoprotein (IDL) and low-density lipoprotein (LDL). ApoB100 has been suggested to play a crucial role in the formation of the atherogenic plaque. Apart from being a biomarker of atherosclerosis, ApoB100 seems to be implicated in the inflammatory process of atherosclerosis per se. In this review, we will focus on the structure, the metabolism, and the function of ApoB100, as well as its role as a predictor biomarker of cardiovascular risk. Moreover, we will elaborate upon the molecular mechanisms regarding the pathophysiology of atherosclerosis, and we will discuss the disorders associated with the APOB gene mutations, and the potential role of various drugs as therapeutic targets.
Collapse
Affiliation(s)
- Dimitris Kounatidis
- Second Department of Internal Medicine, Hippokration General Hospital, School of Medicine, National and Kapodistrian University of Athens, 11527 Athens, Greece;
| | - Natalia G. Vallianou
- Department of Internal Medicine, Evangelismos General Hospital, 10676 Athens, Greece; (F.P.); (E.G.)
| | - Aikaterini Poulaki
- Hematology Unit, Second Department of Internal Medicine, School of Medicine, National and Kapodistrian University of Athens, 11527 Athens, Greece;
| | | | - Fotis Panagopoulos
- Department of Internal Medicine, Evangelismos General Hospital, 10676 Athens, Greece; (F.P.); (E.G.)
| | - Theodora Stratigou
- Department of Endocrinology and Metabolism, Evangelismos General Hospital, 10676 Athens, Greece;
| | - Eleni Geladari
- Department of Internal Medicine, Evangelismos General Hospital, 10676 Athens, Greece; (F.P.); (E.G.)
| | - Irene Karampela
- Second Department of Critical Care, Attikon General University Hospital, Medical School, National and Kapodistrian University of Athens, 12462 Athens, Greece;
| | - Maria Dalamaga
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece;
| |
Collapse
|
19
|
Menekşe TS, Kaçer İ, Hacımustafaoğlu M, Gül M, Ateş C. C-reactive protein to albumin ratio may predict in-hospital mortality in non-ST elevation myocardial infarction. Biomark Med 2024; 18:103-113. [PMID: 38440872 DOI: 10.2217/bmm-2023-0682] [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] [Indexed: 03/06/2024] Open
Abstract
Objective: The authors investigated the value of novel inflammatory markers, systemic immune-inflammation index and C-reactive protein to albumin ratio (CAR), to predict in-hospital mortality in patients with non-ST elevation myocardial infarction (NSTEMI). Materials & methods: A total of 308 patients who underwent percutaneous coronary intervention because of NSTEMI were retrospectively included in the study. Killip classification, Thrombolysis in Myocardial Infarction score, SYNTAX score, and CAR and systemic immune-inflammation index values were calculated. Results: CAR (cutoff: 0.0864; sensitivity: 94.1%; specificity: 40.5%; p = 0.008) and Killip classification (cutoff: 2.5; sensitivity: 64.7%; specificity: 8.9%; p = 0.001) were found to be significantly higher in determining in-hospital mortality. Conclusion: This study revealed that CAR is an inexpensive and significant factor in predicting in-hospital mortality in patients undergoing percutaneous coronary intervention for NSTEMI.
Collapse
Affiliation(s)
- Tuğba S Menekşe
- Department of Emergency Medicine, Aksaray University Training and Research Hospital, Aksaray, Turkey
| | - İlker Kaçer
- Department of Emergency Medicine, Aksaray University Training and Research Hospital, Aksaray, Turkey
| | - Muhammet Hacımustafaoğlu
- Department of Emergency Medicine, Aksaray University Training and Research Hospital, Aksaray, Turkey
| | - Murat Gül
- Department of Cardiology, School of Medicine, Aksaray University, Aksaray, Turkey
| | - Can Ateş
- Department of Biostatistics, Faculty of Medicine, Aksaray University, Aksaray, Turkey
| |
Collapse
|
20
|
He J, Gao Y, Yang C, Guo Y, Liu L, Lu S, He H. Navigating the landscape: Prospects and hurdles in targeting vascular smooth muscle cells for atherosclerosis diagnosis and therapy. J Control Release 2024; 366:261-281. [PMID: 38161032 DOI: 10.1016/j.jconrel.2023.12.047] [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: 09/21/2023] [Revised: 12/02/2023] [Accepted: 12/26/2023] [Indexed: 01/03/2024]
Abstract
Vascular smooth muscle cells (VSMCs) have emerged as pivotal contributors throughout all phases of atherosclerotic plaque development, effectively dispelling prior underestimations of their prevalence and significance. Recent lineage tracing studies have unveiled the clonal nature and remarkable adaptability inherent to VSMCs, thereby illuminating their intricate and multifaceted roles in the context of atherosclerosis. This comprehensive review provides an in-depth exploration of the intricate mechanisms and distinctive characteristics that define VSMCs across various physiological processes, firmly underscoring their paramount importance in shaping the course of atherosclerosis. Furthermore, this review offers a thorough examination of the significant strides made over the past two decades in advancing imaging techniques and therapeutic strategies with a precise focus on targeting VSMCs within atherosclerotic plaques, notably spotlighting meticulously engineered nanoparticles as a promising avenue. We envision the potential of VSMC-targeted nanoparticles, thoughtfully loaded with medications or combination therapies, to effectively mitigate pro-atherogenic VSMC processes. These advancements are poised to contribute significantly to the pivotal objective of modulating VSMC phenotypes and enhancing plaque stability. Moreover, our paper also delves into recent breakthroughs in VSMC-targeted imaging technologies, showcasing their remarkable precision in locating microcalcifications, dynamically monitoring plaque fibrous cap integrity, and assessing the therapeutic efficacy of medical interventions. Lastly, we conscientiously explore the opportunities and challenges inherent in this innovative approach, providing a holistic perspective on the potential of VSMC-targeted strategies in the evolving landscape of atherosclerosis research and treatment.
Collapse
Affiliation(s)
- Jianhua He
- School of Pharmacy, Research Center for Pharmaceutical Preparations, Hubei University of Chinese Medicine, Wuhan 430065, People's Republic of China.
| | - Yu Gao
- School of Pharmacy, China Pharmaceutical University, Nanjing 210009, People's Republic of China
| | - Can Yang
- School of Pharmacy, Research Center for Pharmaceutical Preparations, Hubei University of Chinese Medicine, Wuhan 430065, People's Republic of China
| | - Yujie Guo
- School of Pharmacy, Research Center for Pharmaceutical Preparations, Hubei University of Chinese Medicine, Wuhan 430065, People's Republic of China
| | - Lisha Liu
- School of Pharmacy, China Pharmaceutical University, Nanjing 210009, People's Republic of China.
| | - Shan Lu
- School of Pharmacy, Research Center for Pharmaceutical Preparations, Hubei University of Chinese Medicine, Wuhan 430065, People's Republic of China.
| | - Hongliang He
- State Key Laboratory of Digital Medical Engineering, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Sciences & Medical Engineering, Southeast University, Nanjing 210009, People's Republic of China.
| |
Collapse
|
21
|
Zhao J, Li T, Wang J. Association between psoriasis and dementia: A systematic review. Neurologia 2024; 39:55-62. [PMID: 38161072 DOI: 10.1016/j.nrleng.2023.12.005] [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: 09/30/2020] [Accepted: 12/14/2020] [Indexed: 01/03/2024] Open
Abstract
INTRODUCTION Risk factors for dementia include genetic factors, aging, environmental factors, certain diseases, and unhealthy lifestyle; most types of dementia share a common chronic systemic inflammatory phenotype. Psoriasis is also considered to be a chronic systemic inflammatory disease. It has been suggested that psoriasis may also contribute to the risk of dementia. The aim of this study was to systematically review the literature on the association between psoriasis and dementia. DEVELOPMENT Articles were selected according to the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guidelines. We searched the PubMed and Web of Science databases to identify articles published in peer-reviewed journals and studying the association between psoriasis and dementia. Studies meeting the inclusion criteria were reviewed. We used the Newcastle-Ottawa Scale to assess the quality of each study. After applying the inclusion and exclusion criteria, we included 8 studies for review, 3 of which were found to present a higher risk of bias. Six of the 8 studies supported the hypothesis that prior diagnosis of psoriasis increases the risk of dementia; one study including only a few cases reported that psoriasis decreased the risk of dementia, and one study including relatively young patients found no significant association between psoriasis and the risk of dementia. CONCLUSION Most studies included in this review supported the hypothesis that psoriasis constitutes a risk factor for dementia. However, well-designed stratified cohort studies assessing both psoriasis severity and treatment status are still required to determine the real effect of psoriasis on the risk of dementia and its subtypes.
Collapse
Affiliation(s)
- J Zhao
- Academy of Life Sciences, School of Medicine, Xi'an International University, Xi'an, China.
| | - T Li
- Disease Prevention and Control Section, Shangcai People's Hospital, Shangcai, China
| | - J Wang
- Academy of Life Sciences, School of Medicine, Xi'an International University, Xi'an, China
| |
Collapse
|
22
|
Wu Z, Zhang T, Ma X, Guo S, Zhou Q, Zahoor A, Deng G. Recent advances in anti-inflammatory active components and action mechanisms of natural medicines. Inflammopharmacology 2023; 31:2901-2937. [PMID: 37947913 DOI: 10.1007/s10787-023-01369-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Accepted: 09/16/2023] [Indexed: 11/12/2023]
Abstract
Inflammation is a series of reactions caused by the body's resistance to external biological stimuli. Inflammation affects the occurrence and development of many diseases. Anti-inflammatory drugs have been used widely to treat inflammatory diseases, but long-term use can cause toxic side-effects and affect human functions. As immunomodulators with long-term conditioning effects and no drug residues, natural products are being investigated increasingly for the treatment of inflammatory diseases. In this review, we focus on the inflammatory process and cellular mechanisms in the development of diseases such as inflammatory bowel disease, atherosclerosis, and coronavirus disease-2019. Also, we focus on three signaling pathways (Nuclear factor-kappa B, p38 mitogen-activated protein kinase, Janus kinase/signal transducer and activator of transcription-3) to explain the anti-inflammatory effect of natural products. In addition, we also classified common natural products based on secondary metabolites and explained the association between current bidirectional prediction progress of natural product targets and inflammatory diseases.
Collapse
Affiliation(s)
- Zhimin Wu
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Tao Zhang
- College of Animal Science and Technology, Anhui Agricultural University, Hefei, China
| | - Xiaofei Ma
- College of Veterinary Medicine, Gansu Agriculture University, Lanzhou, China
| | - Shuai Guo
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Qingqing Zhou
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Arshad Zahoor
- College of Veterinary Sciences, The University of Agriculture Peshawar, Peshawar, Pakistan
| | - Ganzhen Deng
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China.
| |
Collapse
|
23
|
Gouda NA, Alshammari SO, Abourehab MAS, Alshammari QA, Elkamhawy A. Therapeutic potential of natural products in inflammation: underlying molecular mechanisms, clinical outcomes, technological advances, and future perspectives. Inflammopharmacology 2023; 31:2857-2883. [PMID: 37950803 DOI: 10.1007/s10787-023-01366-y] [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: 03/20/2023] [Accepted: 10/06/2023] [Indexed: 11/13/2023]
Abstract
Chronic inflammation is a common underlying factor in many major diseases, including heart disease, diabetes, cancer, and autoimmune disorders, and is responsible for up to 60% of all deaths worldwide. Metformin, statins, and corticosteroids, and NSAIDs (non-steroidal anti-inflammatory drugs) are often given as anti-inflammatory pharmaceuticals, however, often have even more debilitating side effects than the illness itself. The natural product-based therapy of inflammation-related diseases has no adverse effects and good beneficial results compared to substitute conventional anti-inflammatory medications. In this review article, we provide a concise overview of present pharmacological treatments, the pathophysiology of inflammation, and the signaling pathways that underlie it. In addition, we focus on the most promising natural products identified as potential anti-inflammatory therapeutic agents. Moreover, preclinical studies and clinical trials evaluating the efficacy of natural products as anti-inflammatory therapeutic agents and their pragmatic applications with promising outcomes are reviewed. In addition, the safety, side effects and technical barriers of natural products are discussed. Furthermore, we also summarized the latest technological advances in the discovery and scientific development of natural products-based medicine.
Collapse
Affiliation(s)
- Noha A Gouda
- College of Pharmacy, Dongguk University-Seoul, Goyang, Gyeonggi, 10326, Republic of Korea
| | - Saud O Alshammari
- Department of Pharmacognosy and Alternative Medicine, Faculty of Pharmacy, Northern Border University, Rafha, 76321, Saudi Arabia
| | - Mohammed A S Abourehab
- Department of Pharmaceutics, College of Pharmacy, Umm Al-Qura University, Makkah, 21955, Saudi Arabia
| | - Qamar A Alshammari
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Northern Border University, Rafha, 76321, Saudi Arabia
| | - Ahmed Elkamhawy
- College of Pharmacy, Dongguk University-Seoul, Goyang, Gyeonggi, 10326, Republic of Korea.
- Department of Pharmaceutical Organic Chemistry, Faculty of Pharmacy, Mansoura University, Mansoura, 35516, Egypt.
| |
Collapse
|
24
|
Lin L, Chen L, Yan J, Chen P, Du J, Zhu J, Yang X, Geng B, Li L, Zeng W. Advances of nanoparticle-mediated diagnostic and theranostic strategies for atherosclerosis. Front Bioeng Biotechnol 2023; 11:1268428. [PMID: 38026849 PMCID: PMC10666776 DOI: 10.3389/fbioe.2023.1268428] [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: 07/28/2023] [Accepted: 10/16/2023] [Indexed: 12/01/2023] Open
Abstract
Atherosclerotic plaque remains the primary cause of morbidity and mortality worldwide. Accurate assessment of the degree of atherosclerotic plaque is critical for predicting the risk of atherosclerotic plaque and monitoring the results after intervention. Compared with traditional technology, the imaging technologies of nanoparticles have distinct advantages and great development prospects in the identification and characterization of vulnerable atherosclerotic plaque. Here, we systematically summarize the latest advances of targeted nanoparticle approaches in the diagnosis of atherosclerotic plaque, including multimodal imaging, fluorescence imaging, photoacoustic imaging, exosome diagnosis, and highlighted the theranostic progress as a new therapeutic strategy. Finally, we discuss the major challenges that need to be addressed for future development and clinical transformation.
Collapse
Affiliation(s)
- Lin Lin
- School of Medicine, Chongqing University, Chongqing, China
- Department of Cell Biology, Third Military Medical University, Chongqing, China
- Jinfeng Laboratory, Chongqing, China
| | - Lin Chen
- Department of Cell Biology, Third Military Medical University, Chongqing, China
| | - Juan Yan
- Department of Cell Biology, Third Military Medical University, Chongqing, China
- Jinfeng Laboratory, Chongqing, China
| | - Peirong Chen
- Department of Cell Biology, Third Military Medical University, Chongqing, China
| | - Jiahui Du
- Department of Cell Biology, Third Military Medical University, Chongqing, China
- Jinfeng Laboratory, Chongqing, China
| | - Junpeng Zhu
- Department of Cell Biology, Third Military Medical University, Chongqing, China
- Jinfeng Laboratory, Chongqing, China
| | - Xinyu Yang
- Department of Cell Biology, Third Military Medical University, Chongqing, China
- Jinfeng Laboratory, Chongqing, China
| | - Boxin Geng
- Department of Cell Biology, Third Military Medical University, Chongqing, China
- Jinfeng Laboratory, Chongqing, China
| | - Lang Li
- Department of Cell Biology, Third Military Medical University, Chongqing, China
- Jinfeng Laboratory, Chongqing, China
| | - Wen Zeng
- School of Medicine, Chongqing University, Chongqing, China
- Department of Cell Biology, Third Military Medical University, Chongqing, China
- Jinfeng Laboratory, Chongqing, China
| |
Collapse
|
25
|
Sun S, Liu F, Fan F, Chen N, Pan X, Wei Z, Zhang Y. Exploring the mechanism of atherosclerosis and the intervention of traditional Chinese medicine combined with mesenchymal stem cells based on inflammatory targets. Heliyon 2023; 9:e22005. [PMID: 38045166 PMCID: PMC10692769 DOI: 10.1016/j.heliyon.2023.e22005] [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: 03/02/2023] [Revised: 11/01/2023] [Accepted: 11/01/2023] [Indexed: 12/05/2023] Open
Abstract
Atherosclerosis (AS) is a chronic inflammatory vascular disease, which is the common pathological basis of cardiovascular and cerebrovascular diseases. The immune inflammatory response throughout the course of AS has been evidenced by studies, in which a large number of immune cells and inflammatory factors play a crucial role in the pathogenesis of AS. The inflammation related to AS is mainly mediated by inflammatory cytokines (IL-1β, IL-6, IL-18, TNF-α, hs-CRP, SAA), inflammatory enzymes (Lp-PLA2, sPLA2-IIA, MMPs), and inflammatory signaling pathways (P38 MAPK signaling pathway, NF-κB signaling pathway, TLR2/4 signaling pathway). It is involved in the pathophysiological process of AS, and the degree of inflammation measured by it can be used to evaluate the risk of progression of AS plaque instability. In recent years, traditional Chinese medicine (TCM) has shown the advantage of minimal side effects in immune regulation and has made some progress in the prevention and treatment of AS. Mesenchymal stem cells (MSCs), as self-renewal, highly differentiated, and pluripotent stem cells with anti-inflammatory properties and immune regulation, have been widely used for AS treatment. They also play an important inflammation-immune regulatory function in AS. Notably, in terms of regulating immune cells and inflammatory factors, compared with TCM and its compound, the combination therapy has obvious anti-inflammatory advantages over the use of MSCs alone. It is an important means to further improve the efficacy of AS and provides a new way for the prevention and treatment of AS.
Collapse
Affiliation(s)
- Shibiao Sun
- Henan University of Chinese Medicine, Zhengzhou 450000, China
| | - Feixiang Liu
- The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou 450000, China
| | - Feiyan Fan
- Henan University of Chinese Medicine, Zhengzhou 450000, China
| | - Na Chen
- Henan University of Chinese Medicine, Zhengzhou 450000, China
| | - Xiaolong Pan
- Henan University of Chinese Medicine, Zhengzhou 450000, China
| | - Zhihui Wei
- Henan University of Chinese Medicine, Zhengzhou 450000, China
| | - Yunke Zhang
- Henan University of Chinese Medicine, Zhengzhou 450000, China
- The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou 450000, China
| |
Collapse
|
26
|
Karnewar S, Karnewar V, Deaton R, Shankman LS, Benavente ED, Williams CM, Bradley X, Alencar GF, Bulut GB, Kirmani S, Baylis RA, Zunder ER, den Ruijter HM, Pasterkamp G, Owens GK. IL-1β inhibition partially negates the beneficial effects of diet-induced lipid lowering. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.10.13.562255. [PMID: 37873280 PMCID: PMC10592822 DOI: 10.1101/2023.10.13.562255] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/25/2023]
Abstract
Background Thromboembolic events secondary to rupture or erosion of advanced atherosclerotic lesions are the leading cause of death in the world. The most common and effective means to reduce these major adverse cardiovascular events (MACE), including myocardial infarction (MI) and stroke, is aggressive lipid lowering via a combination of drugs and dietary modifications. However, little is known regarding the effects of reducing dietary lipids on the composition and stability of advanced atherosclerotic lesions, the mechanisms that regulate these processes, and what therapeutic approaches might augment the benefits of lipid lowering. Methods Smooth muscle cell (SMC)-lineage tracing Apoe-/- mice were fed a Western diet (WD) for 18 weeks and then switched to a low-fat chow diet for 12 weeks. We assessed lesion size and remodeling indices, as well as the cellular composition of aortic and brachiocephalic artery (BCA) lesions, indices of plaque stability, overall plaque burden, and phenotypic transitions of SMC, and other lesion cells by SMC-lineage tracing combined with scRNA-seq, CyTOF, and immunostaining plus high resolution confocal microscopic z-stack analysis. In addition, to determine if treatment with a potent inhibitor of inflammation could augment the benefits of chow diet-induced reductions in LDL-cholesterol, SMC-lineage tracing Apoe-/- mice were fed a WD for 18 weeks and then chow diet for 12 weeks prior to treating them with an IL-1β or control antibody (Ab) for 8-weeks. Results Lipid-lowering by switching Apoe-/- mice from a WD to a chow diet reduced LDL-cholesterol levels by 70% and resulted in multiple beneficial effects including reduced overall aortic plaque burden as well as reduced intraplaque hemorrhage and necrotic core area. However, contrary to expectations, IL-1β Ab treatment resulted in multiple detrimental changes including increased plaque burden, BCA lesion size, as well as increased cholesterol crystal accumulation, intra-plaque hemorrhage, necrotic core area, and senescence as compared to IgG control Ab treated mice. Furthermore, IL-1β Ab treatment upregulated neutrophil degranulation pathways but down-regulated SMC extracellular matrix pathways likely important for the protective fibrous cap. Conclusions Taken together, IL-1β appears to be required for chow diet-induced reductions in plaque burden and increases in multiple indices of plaque stability.
Collapse
Affiliation(s)
- Santosh Karnewar
- Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville, USA
| | - Vaishnavi Karnewar
- Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville, USA
| | - Rebecca Deaton
- Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville, USA
| | - Laura S. Shankman
- Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville, USA
| | - Ernest D. Benavente
- Laboratory of Experimental Cardiology, University Medical Center Utrecht, Utrecht University, the Netherlands
| | - Corey M. Williams
- Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville, USA
| | - Xenia Bradley
- Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville, USA
| | - Gabriel F. Alencar
- Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville, USA
| | - Gamze B. Bulut
- Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville, USA
| | - Sara Kirmani
- Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville, USA
| | - Richard A. Baylis
- Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville, USA
| | - Eli R. Zunder
- Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville, USA
| | - Hester M. den Ruijter
- Laboratory of Experimental Cardiology, University Medical Center Utrecht, Utrecht University, the Netherlands
| | - Gerard Pasterkamp
- Laboratory of Experimental Cardiology, University Medical Center Utrecht, Utrecht University, the Netherlands
| | - Gary K. Owens
- Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville, USA
| |
Collapse
|
27
|
Catană MG, Popențiu IA, Văleanu M, Roman-Filip C, Mihăilă RG. IL-1 Beta-A Biomarker for Ischemic Stroke Prognosis and Atherosclerotic Lesions of the Internal Carotid Artery. MEDICINA (KAUNAS, LITHUANIA) 2023; 59:1790. [PMID: 37893508 PMCID: PMC10608497 DOI: 10.3390/medicina59101790] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/26/2023] [Revised: 09/26/2023] [Accepted: 10/06/2023] [Indexed: 10/29/2023]
Abstract
Background and Objectives: Stroke is a leading cause of mortality and morbidity worldwide. Treatment of this pathology is still under development and its risk factors remain to be determined. Therefore, we aim to determine the role of interleukin-1 beta in atherosclerotic lesions of the internal carotid artery as a risk factor for stroke and the role of this biomarker in stroke prognosis. Materials and Methods: This study enrolled 56 patients diagnosed with ischemic stroke in the anterior vascular territory (AVT) and posterior vascular territory (PVT). All the patients had venous blood collected at admission and 7 days after the onset of the cerebral ischemia in order to determine the plasma concentration of interleukin-1 beta. At the same time, an extracranial carotid ultrasound was performed. Results: The interleukin-1 beta collected at admission was positively correlated with the NIHSS at admission (Pearson index 0.424), and both measurements were correlated with carotid stenosis (Spearmen correlation index of 0.529 and 0.653, respectively). Conclusions: Interleukin-1 beta could be a reliable biomarker for stroke prognosis and the development of atherosclerotic lesions of the internal carotid.
Collapse
Affiliation(s)
- Maria-Gabriela Catană
- Faculty of Medicine, Lucian Blaga University of Sibiu, Izvorului Street, 550169 Sibiu, Romania
- Neurology Department, Emergency County Clinical Hospital Sibiu, Corneliu Coposu bvd, 550245 Sibiu, Romania
| | - Ioan-Adrian Popențiu
- Faculty of Medicine, Lucian Blaga University of Sibiu, Izvorului Street, 550169 Sibiu, Romania
- Department of General Surgery, “Alexandru Augustin” Military Emergency Hospital, Victoriei bvd, 550024 Sibiu, Romania
| | - Mădălina Văleanu
- Department of Medical Informatics and Biostatistics, University of Medicine and Pharmacy “Iuliu Hatieganu” Cluj-Napoca, 7 Horea Street, 400174 Cluj-Napoca, Romania
| | - Corina Roman-Filip
- Faculty of Medicine, Lucian Blaga University of Sibiu, Izvorului Street, 550169 Sibiu, Romania
- Neurology Department, Emergency County Clinical Hospital Sibiu, Corneliu Coposu bvd, 550245 Sibiu, Romania
| | - Romeo-Gabriel Mihăilă
- Faculty of Medicine, Lucian Blaga University of Sibiu, Izvorului Street, 550169 Sibiu, Romania
- Hematology Department, Emergency County Clinical Hospital Sibiu, Sibiu Corneliu Coposu bvd, 550245 Sibiu, Romania
| |
Collapse
|
28
|
Ji W, Zhang Y, Deng Y, Li C, Kankala RK, Chen A. Nature-inspired nanocarriers for improving drug therapy of atherosclerosis. Regen Biomater 2023; 10:rbad069. [PMID: 37641591 PMCID: PMC10460486 DOI: 10.1093/rb/rbad069] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Revised: 07/22/2023] [Accepted: 08/02/2023] [Indexed: 08/31/2023] Open
Abstract
Atherosclerosis (AS) has emerged as one of the prevalent arterial vascular diseases characterized by plaque and inflammation, primarily causing disability and mortality globally. Drug therapy remains the main treatment for AS. However, a series of obstacles hinder effective drug delivery. Nature, from natural micro-/nano-structural biological particles like natural cells and extracellular vesicles to the distinctions between the normal and pathological microenvironment, offers compelling solutions for efficient drug delivery. Nature-inspired nanocarriers of synthetic stimulus-responsive materials and natural components, such as lipids, proteins and membrane structures, have emerged as promising candidates for fulfilling drug delivery needs. These nanocarriers offer several advantages, including prolonged blood circulation, targeted plaque delivery, targeted specific cells delivery and controlled drug release at the action site. In this review, we discuss the nature-inspired nanocarriers which leverage the natural properties of cells or the microenvironment to improve atherosclerotic drug therapy. Finally, we provide an overview of the challenges and opportunities of applying these innovative nature-inspired nanocarriers.
Collapse
Affiliation(s)
- Weihong Ji
- Institute of Biomaterials and Tissue Engineering, Huaqiao University, Xiamen, Fujian 361021, PR China
- Fujian Provincial Key Laboratory of Biochemical Technology, Huaqiao University, Xiamen, Fujian 361021, PR China
| | - Yuanxing Zhang
- The Institute of Forensic Science, Xiamen Public Security Bureau, Xiamen, Fujian 361104, PR China
| | - Yuanru Deng
- Institute of Biomaterials and Tissue Engineering, Huaqiao University, Xiamen, Fujian 361021, PR China
- Fujian Provincial Key Laboratory of Biochemical Technology, Huaqiao University, Xiamen, Fujian 361021, PR China
| | - Changyong Li
- Institute of Biomaterials and Tissue Engineering, Huaqiao University, Xiamen, Fujian 361021, PR China
- Fujian Provincial Key Laboratory of Biochemical Technology, Huaqiao University, Xiamen, Fujian 361021, PR China
| | - Ranjith Kumar Kankala
- Institute of Biomaterials and Tissue Engineering, Huaqiao University, Xiamen, Fujian 361021, PR China
- Fujian Provincial Key Laboratory of Biochemical Technology, Huaqiao University, Xiamen, Fujian 361021, PR China
| | - Aizheng Chen
- Institute of Biomaterials and Tissue Engineering, Huaqiao University, Xiamen, Fujian 361021, PR China
- Fujian Provincial Key Laboratory of Biochemical Technology, Huaqiao University, Xiamen, Fujian 361021, PR China
| |
Collapse
|
29
|
Bradley NA, Walter A, Wilson A, Siddiqui T, Roxburgh CSD, McMillan DC, Guthrie GJK. The prognostic value of preoperative systemic inflammation-based scoring in patients undergoing endovascular repair of abdominal aortic aneurysm. J Vasc Surg 2023; 78:362-369.e2. [PMID: 37086821 DOI: 10.1016/j.jvs.2023.04.018] [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: 02/24/2023] [Revised: 04/04/2023] [Accepted: 04/11/2023] [Indexed: 04/24/2023]
Abstract
OBJECTIVE Abdominal aortic aneurysm (AAA) is a common condition that is predominantly managed in the United Kingdom by endovascular aneurysm repair (EVAR). Activation of the systemic inflammatory response (SIR) appears to offer prognostic value in patients with vascular disease. The present study examines the relationship between the SIR and survival in patients undergoing standard and complex endovascular aneurysm repair (EVAR and fenestrated/branched [F/B]-EVAR). METHODS Consecutive patients undergoing elective EVAR and F/B-EVAR were retrospectively identified from three tertiary vascular centers over a 5-year period. Neutrophil:lymphocyte ratio and modified Glasgow Prognostic Score were calculated from preoperative blood results and combined into the systemic inflammatory grade (SIG). The primary outcome was all-cause mortality during the follow-up period, which was compared between subgroups of SIGs. RESULTS There were 506 patients included in the final study, with a median follow-up of 68.0 months (interquartile range, 27.3 months), and there were 163 deaths during the follow-up period. Mean survival in the SIG 0 vs SIG 1 vs SIG 2 vs SIG 3 vs SIG 4 subgroups was 80.7 months (95% confidence interval [CI], 76.5-85.0 months) vs 78.7 months (95% CI, 72.7-84.7 months) vs 61.0 months (95% CI, 51.1-70.8 months) vs 65.1 months (95% CI, 45.0-85.2 months) vs 54.9 months (95% CI, 34.4-75.3 months) (P < .05). In the entire cohort, age (P < .001), body mass index (P < .05), high creatinine (P < .05), and SIG (P < .05) were associated with survival on univariate analysis, with retained independent association for age (hazard ratio, 1.72; 95% CI, 1.29-2.31; P < .001) and SIG (hazard ratio, 1.20; 95% CI, 1.02-1.40; P < .05) on multivariate analysis. Increasing SIG (area under the curve, 0.68; 95% CI, 0.58-0.78; P < .01) predicted 1-year mortality. CONCLUSIONS Markers of the SIR such the SIG may be used to identify patients at higher risk of adverse outcome in patients undergoing EVAR and F/B-EVAR for abdominal aortic aneurysms. These findings warrant further investigation in large prospective cohort studies.
Collapse
Affiliation(s)
- Nicholas A Bradley
- Academic Unit of Surgery, University of Glasgow, Glasgow, United Kingdom.
| | - Amy Walter
- Department of Vascular Surgery, NHS Tayside, Dundee, United Kingdom
| | - Alasdair Wilson
- Department of Vascular Surgery, NHS Grampian, Aberdeen, United Kingdom
| | - Tamim Siddiqui
- Department of Vascular Surgery, NHS Lanarkshire, Glasgow, United Kingdom
| | | | - Donald C McMillan
- Academic Unit of Surgery, University of Glasgow, Glasgow, United Kingdom
| | - Graeme J K Guthrie
- Academic Unit of Surgery, University of Glasgow, Glasgow, United Kingdom; Department of Vascular Surgery, NHS Tayside, Dundee, United Kingdom
| |
Collapse
|
30
|
Bradley NA, Walter A, Dolan R, Wilson A, Siddiqui T, Roxburgh CS, McMillan DC, Guthrie GJ. Evaluation of the prognostic value of computed tomography-derived body composition in patients undergoing endovascular aneurysm repair. J Cachexia Sarcopenia Muscle 2023; 14:1836-1847. [PMID: 37221439 PMCID: PMC10401537 DOI: 10.1002/jcsm.13262] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Revised: 01/23/2023] [Accepted: 04/24/2023] [Indexed: 05/25/2023] Open
Abstract
BACKGROUND Endovascular aneurysm repair (EVAR) is the most common mode of repair of abdominal aortic aneurysms (AAA) in the UK. EVAR ranges from standard infrarenal repair to complex fenestrated and branched EVAR (F/B-EVAR). Sarcopenia is defined by lower muscle mass and function, which is associated with inferior perioperative outcomes. Computed tomography-derived body composition analysis offers prognostic value in patients with cancer. Several authors have evaluated the role of body composition analysis in predicting outcomes in patients undergoing EVAR; however, the evidence base is limited by heterogeneous methodology. METHODS Six hundred seventy-four consecutive patients (58 (8.6%) female, mean (SD) age 74.4 (6.8) years) undergoing EVAR and F/B-EVAR at three large tertiary centres were retrospectively recruited. Subcutaneous and visceral fat indices (SFI and VFI), psoas and skeletal muscle indices, and skeletal muscle density were measured at the L3 vertebral level from pre-operative computed tomographies. The maximally selected rank statistic technique was used to define optimal thresholds to predict mortality. RESULTS There were 191 deaths during the median follow-up period of 60.0 months. Mean (95% CI) survival in the low SMI versus high SMI subgroups was 62.6 (58.5-66.7) versus 82.0 (78.7-85.3) months (P < 0.001). Mean (95% CI) survival in the low SFI versus high SFI subgroups was 56.4 (48.2-64.7) versus 77.1 (74.2-80.1) months (P < 0.001). One-year mortality in the low SMI versus high SMI subgroups was 10% versus 3% (P < 0.001). Low SMI was associated with increased odds of one-year mortality (OR 3.19, 95% CI 1.60-6.34, P < 0.001). Five-year mortality in the low SMI versus high SMI subgroups was 55% versus 28% (P < 0.001). Low SMI was associated with increased odds of five-year mortality (OR 1.54, 95% CI 1.11-2.14, P < 0.01). On multivariate analysis of all patients, low SFI (HR 1.90, 95% CI 1.30-2.76, P < 0.001) and low SMI (HR 1.88, 95% CI 1.34-2.63, P < 0.001) were associated with poorer survival. On multivariate analysis of asymptomatic AAA patients, low SFI (HR 1.54, 95% CI 1.01-2.35, P < 0.05) and low SMI (HR 1.71, 95% CI 1.20-2.42, P < 0.01) were associated with poorer survival. CONCLUSIONS Low SMI and SFI are associated with poorer long-term survival following EVAR and F/B-EVAR. The relationship between body composition and prognosis requires further evaluation, and external validation of the thresholds proposed in patients with AAA is required.
Collapse
|
31
|
Zhang W, Zhao J, Deng L, Ishimwe N, Pauli J, Wu W, Shan S, Kempf W, Ballantyne MD, Kim D, Lyu Q, Bennett M, Rodor J, Turner AW, Lu YW, Gao P, Choi M, Warthi G, Kim HW, Barroso MM, Bryant WB, Miller CL, Weintraub NL, Maegdefessel L, Miano JM, Baker AH, Long X. INKILN is a Novel Long Noncoding RNA Promoting Vascular Smooth Muscle Inflammation via Scaffolding MKL1 and USP10. Circulation 2023; 148:47-67. [PMID: 37199168 PMCID: PMC10330325 DOI: 10.1161/circulationaha.123.063760] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Accepted: 04/14/2023] [Indexed: 05/19/2023]
Abstract
BACKGROUND Activation of vascular smooth muscle cell (VSMC) inflammation is vital to initiate vascular disease. The role of human-specific long noncoding RNAs in VSMC inflammation is poorly understood. METHODS Bulk RNA sequencing in differentiated human VSMCs revealed a novel human-specific long noncoding RNA called inflammatory MKL1 (megakaryoblastic leukemia 1) interacting long noncoding RNA (INKILN). INKILN expression was assessed in multiple in vitro and ex vivo models of VSMC phenotypic modulation as well as human atherosclerosis and abdominal aortic aneurysm. The transcriptional regulation of INKILN was verified through luciferase reporter and chromatin immunoprecipitation assays. Loss-of-function and gain-of-function studies and multiple RNA-protein and protein-protein interaction assays were used to uncover a mechanistic role of INKILN in the VSMC proinflammatory gene program. Bacterial artificial chromosome transgenic mice were used to study INKILN expression and function in ligation injury-induced neointimal formation. RESULTS INKILN expression is downregulated in contractile VSMCs and induced in human atherosclerosis and abdominal aortic aneurysm. INKILN is transcriptionally activated by the p65 pathway, partially through a predicted NF-κB (nuclear factor kappa B) site within its proximal promoter. INKILN activates proinflammatory gene expression in cultured human VSMCs and ex vivo cultured vessels. INKILN physically interacts with and stabilizes MKL1, a key activator of VSMC inflammation through the p65/NF-κB pathway. INKILN depletion blocks interleukin-1β-induced nuclear localization of both p65 and MKL1. Knockdown of INKILN abolishes the physical interaction between p65 and MKL1 and the luciferase activity of an NF-κB reporter. Furthermore, INKILN knockdown enhances MKL1 ubiquitination through reduced physical interaction with the deubiquitinating enzyme USP10 (ubiquitin-specific peptidase 10). INKILN is induced in injured carotid arteries and exacerbates ligation injury-induced neointimal formation in bacterial artificial chromosome transgenic mice. CONCLUSIONS These findings elucidate an important pathway of VSMC inflammation involving an INKILN/MKL1/USP10 regulatory axis. Human bacterial artificial chromosome transgenic mice offer a novel and physiologically relevant approach for investigating human-specific long noncoding RNAs under vascular disease conditions.
Collapse
Affiliation(s)
- Wei Zhang
- Vascular Biology Center, Medical College of Georgia at Augusta University, Augusta, GA, USA
| | - Jinjing Zhao
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY, USA
| | - Lin Deng
- Centre for Cardiovascular Science University of Edinburgh, Edinburgh, Scotland
| | - Nestor Ishimwe
- Vascular Biology Center, Medical College of Georgia at Augusta University, Augusta, GA, USA
| | - Jessica Pauli
- Department for Vascular and Endovascular Surgery, Klinikum rechts der Isar, Technical University Munich, Germany
| | - Wen Wu
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY, USA
| | - Shengshuai Shan
- Vascular Biology Center, Medical College of Georgia at Augusta University, Augusta, GA, USA
| | - Wolfgang Kempf
- Department for Vascular and Endovascular Surgery, Klinikum rechts der Isar, Technical University Munich, Germany
| | | | - David Kim
- Vascular Biology Center, Medical College of Georgia at Augusta University, Augusta, GA, USA
| | - Qing Lyu
- Vascular Biology Center, Medical College of Georgia at Augusta University, Augusta, GA, USA
| | - Matthew Bennett
- Centre for Cardiovascular Science University of Edinburgh, Edinburgh, Scotland
| | - Julie Rodor
- Centre for Cardiovascular Science University of Edinburgh, Edinburgh, Scotland
| | - Adam W. Turner
- Center for Public Health Genomics, University of Virginia, Charlottesville, VA, USA
| | - Yao Wei Lu
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY, USA
| | - Ping Gao
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY, USA
| | - Mihyun Choi
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY, USA
| | - Ganesh Warthi
- Vascular Biology Center, Medical College of Georgia at Augusta University, Augusta, GA, USA
| | - Ha Won Kim
- Vascular Biology Center, Medical College of Georgia at Augusta University, Augusta, GA, USA
| | - Margarida M Barroso
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY, USA
| | - William B. Bryant
- Vascular Biology Center, Medical College of Georgia at Augusta University, Augusta, GA, USA
| | - Clint L. Miller
- Center for Public Health Genomics, University of Virginia, Charlottesville, VA, USA
- Department of Biochemistry and Molecular Genetics, University of Virginia, Charlottesville, VA, USA
| | - Neal L. Weintraub
- Vascular Biology Center, Medical College of Georgia at Augusta University, Augusta, GA, USA
| | - Lars Maegdefessel
- Department for Vascular and Endovascular Surgery, Klinikum rechts der Isar, Technical University Munich, Germany
- German Center for Cardiovascular Research (DZHK, partner site Munich), Germany
- Department of Medicine, Karolinska Institute, Stockholm, Sweden
| | - Joseph M. Miano
- Vascular Biology Center, Medical College of Georgia at Augusta University, Augusta, GA, USA
| | - Andrew H Baker
- Centre for Cardiovascular Science University of Edinburgh, Edinburgh, Scotland
| | - Xiaochun Long
- Vascular Biology Center, Medical College of Georgia at Augusta University, Augusta, GA, USA
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY, USA
| |
Collapse
|
32
|
Mewborn EK, Wright DB, Wicks MN. Clinical utility of inflammatory and genetic biomarkers for cardiovascular disease prevention, categorization, and treatment. J Am Assoc Nurse Pract 2023; 35:404-412. [PMID: 37260280 DOI: 10.1097/jxx.0000000000000901] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Accepted: 05/09/2023] [Indexed: 06/02/2023]
Abstract
ABSTRACT The complex interplay of increased atherogenic lipoproteins, inflammation, and immune activation hallmarks the pathogenesis of atherosclerotic cardiovascular disease (ASCVD). Atherosclerotic cardiovascular disease remains a leading cause of death, yet risk estimator tools lack comprehensiveness for genetic/inflammatory biomarkers associated with ASCVD. Unexplained ASCVD risk necessitates a better understanding of primary, secondary, and tertiary prevention variables. This article discusses the clinical utility of genetic and inflammatory biomarkers for ASCVD risk prediction, management, treatment, and recategorization into primary, secondary, and tertiary prevention. Furthermore, nurse practitioners (NPs) should use a ternary prevention classification system instead of the current binary system to mitigate risk in the large group of patients with subclinical ASCVD. High-sensitivity C-reactive protein (hs-CRP)-linearly associated with ASCVD-and lipoprotein-associated phospholipase-A 2 (Lp-PLA 2 ) and myeloperoxidase (MPO), both associated with plaque vulnerability/rupture, are inflammatory biomarkers. Elevated hs-CRP, MPO, and Lp-PLA 2 treatment requires addressing root causes of elevation (e.g., obesity, insulin resistance, tobacco use, gingival disease, and chronic autoimmune/infectious conditions). In addition, haptoglobin (Hp) phenotype determines the antioxidant potential of Hp. Haptoglobin phenotype, a root cause of ASCVD, is a one-time test. Individuals with Hp 2-2 should adopt a gluten-free diet to reduce endothelial and intestinal inflammation. Nurse practitioners should use stricter glycemic goals (hemoglobin A1c ≤6.5%) and add daily vitamin E if this group has type 2 diabetes. Genetic/inflammatory biomarkers should be used to better predict ASCVD risk and tailor primary, secondary, and tertiary prevention treatment. Clinical use of these biomarkers reaches beyond the standard of care to reduce residual ASCVD risk.
Collapse
Affiliation(s)
- Emily K Mewborn
- Baptist Medical Group, Collierville, Tennessee
- The University of Tennessee Health Science Center, Memphis, Tennessee
| | | | - Mona N Wicks
- Health Promotion and Disease Prevention, The University of Tennessee Health Science Center, Memphis, Tennessee
| |
Collapse
|
33
|
Rocha VZ, Rached FH, Miname MH. Insights into the Role of Inflammation in the Management of Atherosclerosis. J Inflamm Res 2023; 16:2223-2239. [PMID: 37250107 PMCID: PMC10225146 DOI: 10.2147/jir.s276982] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2022] [Accepted: 03/28/2023] [Indexed: 05/31/2023] Open
Abstract
Atherosclerosis is the biological basis of ischemic heart disease and ischemic stroke, the leading causes of death in the world. After decades of studies, the understanding of atherosclerosis has evolved dramatically, and inflammation has been recognized as one of the most relevant pillars in all phases of atherosclerotic disease. Nevertheless, only recently, the trial CANTOS, and subsequent outcome studies with colchicine, finally provided proof-of-concept evidence that anti-inflammatory therapies were able to reduce cardiovascular events with no influence on lipid levels. These landmark studies inaugurated an era of clinical and pre-clinical studies of immunomodulatory strategies focused on reduction of cardiovascular risk. Although there are promising results in the field, selection of the most appropriate immunomodulatory therapy and identification of patients who could benefit the most, are still enormous challenges. Further research is imperative before we can finally advance towards regular use of anti-inflammatory agents to reduce atherosclerotic events in our clinical practice.
Collapse
Affiliation(s)
- Viviane Zorzanelli Rocha
- Cardiopneumology Department at the Heart Institute (InCor), University of São Paulo Medical School Hospital (FMUSP), Sao Paulo, Brazil
- Fleury Medicina e Saúde, Grupo Fleury, São Paulo, SP, Brazil
| | - Fabiana Hanna Rached
- Cardiopneumology Department at the Heart Institute (InCor), University of São Paulo Medical School Hospital (FMUSP), Sao Paulo, Brazil
- Department of Cardiology, Hospital Israelita Albert Einstein, São Paulo, Brazil
| | - Marcio Hiroshi Miname
- Cardiopneumology Department at the Heart Institute (InCor), University of São Paulo Medical School Hospital (FMUSP), Sao Paulo, Brazil
| |
Collapse
|
34
|
Chen Q, Che M, Shen W, Shao L, Yu H, Zhou J. Comparison of the Early Warning Effects of Novel Inflammatory Markers SIRI, NLR, and LMR in the Inhibition of Carotid Atherosclerosis by Testosterone in Middle-Aged and Elderly Han Chinese Men in the Real World: A Small Sample Clinical Observational Study. Am J Mens Health 2023; 17:15579883231171462. [PMID: 37183913 DOI: 10.1177/15579883231171462] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/16/2023] Open
Abstract
The purpose of this study was to explore and compare the relationship among serum testosterone, systemic inflammatory response index (SIRI), lymphocyte-to-monocyte ratio (LMR) neutrophil-lymphocyte ratio (NLR), and carotid atherosclerosis in middle-aged and elderly men of Han nationality in the real world. With reference to the inclusion criteria, 89 middle-aged and elderly Han male patients were finally selected. Local weighted regression (LOESS) and multivariate logistic regression models were used to explore the independent correlation between serum testosterone, new inflammatory markers, and atherosclerosis. The diagnostic value of related indexes was evaluated by the receiver working curve characteristic curve (ROC), and the best critical value of testosterone and related inflammatory indexes was discussed. In the LOESS model, bioavailable testosterone (BT), free testosterone (FT), total testosterone (TT) and SIRI, NLR, LMR, and atherosclerosis were significantly correlated. After adjusting for confounding factors, BT, FT, TT, and LMR were negatively correlated with atherosclerosis (odds ratio [OR] < 1, p < .05), and SIRI and NLR were positively associated with atherosclerosis (OR > 1, p < .05). According to the ROC curve results, the area under the curve (AUG) of BT is 0.870, and the optimal threshold point is 4.875. The AUG of SIRI is 0.864, and the best threshold point is 0.769. Low testosterone and high inflammatory levels are closely related to atherosclerosis. Testosterone (TT, FT, and BT) and new inflammatory markers, SIRI, NLR, and LMR, are associated with carotid atherosclerosis in middle-aged and elderly men.
Collapse
Affiliation(s)
- Qinhao Chen
- Department of Geriatrics, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
- Department of Oncology, Provincial Hospital Affiliated to Anhui Medical University, Hefei, Anhui, China
| | - Mingzhu Che
- Department of Preventive Medicine, School of Public Health and Management, Wenzhou Medical University, Wenzhou, China
| | - Wei Shen
- Department of International Medical, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| | - Lijie Shao
- Department of Geriatrics, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| | - Haiyang Yu
- Department of Oncology, Provincial Hospital Affiliated to Anhui Medical University, Hefei, Anhui, China
| | - Jian Zhou
- Department of Geriatrics, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
- Department of Geriatrics, Wannan Medical College, Wuhu, Anhui, China
| |
Collapse
|
35
|
Schwarz N, Fernando S, Chen YC, Salagaras T, Rao SR, Liyanage S, Williamson AE, Toledo-Flores D, Dimasi C, Sargeant TJ, Manavis J, Eddy E, Kanellakis P, Thompson PL, Tan JTM, Snel MF, Bursill CA, Nicholls SJ, Peter K, Psaltis PJ. Colchicine exerts anti-atherosclerotic and -plaque-stabilizing effects targeting foam cell formation. FASEB J 2023; 37:e22846. [PMID: 36856983 DOI: 10.1096/fj.202201469r] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Revised: 02/02/2023] [Accepted: 02/15/2023] [Indexed: 03/02/2023]
Abstract
Colchicine is a broad-acting anti-inflammatory agent that has attracted interest for repurposing in atherosclerotic cardiovascular disease. Here, we studied its ability at a human equivalent dose of 0.5 mg/day to modify plaque formation and composition in murine atherosclerosis and investigated its actions on macrophage responses to atherogenic stimuli in vitro. In atherosclerosis induced by high-cholesterol diet, Apoe-/- mice treated with colchicine had 50% reduction in aortic oil Red O+ plaque area compared to saline control (p = .001) and lower oil Red O+ staining of aortic sinus lesions (p = .03). In vitro, addition of 10 nM colchicine inhibited foam cell formation from murine and human macrophages after treatment with oxidized LDL (ox-LDL). Mechanistically, colchicine downregulated glycosylation and surface expression of the ox-LDL uptake receptor, CD36, and reduced CD36+ staining in aortic sinus plaques. It also decreased macrophage uptake of cholesterol crystals, resulting in lower intracellular lysosomal activity, inhibition of the NLRP3 inflammasome, and reduced secretion of IL-1β and IL-18. Colchicine's anti-atherosclerotic actions were accentuated in a mouse model of unstable plaque induced by carotid artery tandem stenosis surgery, where it decreased lesion size by 48% (p = .01), reduced lipid (p = .006) and necrotic core area (p = .007), increased collagen content and cap-to-necrotic core ratio (p = .05), and attenuated plaque neutrophil extracellular traps (p < .001). At low dose, colchicine's effects were not accompanied by the evidence of microtubule depolymerization. Together, these results show that colchicine exerts anti-atherosclerotic and plaque-stabilizing effects at low dose by inhibiting foam cell formation and cholesterol crystal-induced inflammation. This provides a new framework to support its repurposing for atherosclerotic cardiovascular disease.
Collapse
Affiliation(s)
- Nisha Schwarz
- Vascular Research Centre, Heart and Vascular Program, Lifelong Health Theme, South Australian Health and Medical Research Institute, Adelaide, South Australia, Australia
| | - Sanuja Fernando
- Vascular Research Centre, Heart and Vascular Program, Lifelong Health Theme, South Australian Health and Medical Research Institute, Adelaide, South Australia, Australia.,Adelaide Medical School, University of Adelaide, Adelaide, South Australia, Australia
| | - Yung-Chih Chen
- Atherothrombosis and Vascular Biology, Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
| | - Thalia Salagaras
- Vascular Research Centre, Heart and Vascular Program, Lifelong Health Theme, South Australian Health and Medical Research Institute, Adelaide, South Australia, Australia
| | - Sushma R Rao
- Proteomics, Metabolomics and MS-Imaging Facility, South Australian Health and Medical Research Institute, Adelaide, South Australia, Australia
| | - Sanuri Liyanage
- Vascular Research Centre, Heart and Vascular Program, Lifelong Health Theme, South Australian Health and Medical Research Institute, Adelaide, South Australia, Australia.,Adelaide Medical School, University of Adelaide, Adelaide, South Australia, Australia
| | - Anna E Williamson
- Vascular Research Centre, Heart and Vascular Program, Lifelong Health Theme, South Australian Health and Medical Research Institute, Adelaide, South Australia, Australia.,Adelaide Medical School, University of Adelaide, Adelaide, South Australia, Australia
| | - Deborah Toledo-Flores
- Vascular Research Centre, Heart and Vascular Program, Lifelong Health Theme, South Australian Health and Medical Research Institute, Adelaide, South Australia, Australia
| | - Catherine Dimasi
- Vascular Research Centre, Heart and Vascular Program, Lifelong Health Theme, South Australian Health and Medical Research Institute, Adelaide, South Australia, Australia
| | - Timothy J Sargeant
- Lifelong Health in Ageing, Hopwood Centre for Neurobiology, Lifelong Health Theme, South Australian Health and Medical Research Institute, Adelaide, South Australia, Australia
| | - Jim Manavis
- Adelaide Medical School, University of Adelaide, Adelaide, South Australia, Australia
| | - Eleanor Eddy
- Atherothrombosis and Vascular Biology, Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
| | - Peter Kanellakis
- Atherothrombosis and Vascular Biology, Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
| | - Peter L Thompson
- Faculty of Health and Medical Sciences, Internal Medicine, The University of Western Australia, Perth, Western Australia, Australia
| | - Joanne T M Tan
- Vascular Research Centre, Heart and Vascular Program, Lifelong Health Theme, South Australian Health and Medical Research Institute, Adelaide, South Australia, Australia.,Adelaide Medical School, University of Adelaide, Adelaide, South Australia, Australia
| | - Marten F Snel
- Proteomics, Metabolomics and MS-Imaging Facility, South Australian Health and Medical Research Institute, Adelaide, South Australia, Australia
| | - Christina A Bursill
- Vascular Research Centre, Heart and Vascular Program, Lifelong Health Theme, South Australian Health and Medical Research Institute, Adelaide, South Australia, Australia.,Adelaide Medical School, University of Adelaide, Adelaide, South Australia, Australia
| | - Stephen J Nicholls
- Monash Cardiovascular Research Centre, Monash University, Melbourne, Victoria, Australia
| | - Karlheinz Peter
- Atherothrombosis and Vascular Biology, Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
| | - Peter J Psaltis
- Vascular Research Centre, Heart and Vascular Program, Lifelong Health Theme, South Australian Health and Medical Research Institute, Adelaide, South Australia, Australia.,Adelaide Medical School, University of Adelaide, Adelaide, South Australia, Australia
| |
Collapse
|
36
|
Avagimyan A, Gvianishvili T, Gogiashvili L, Kakturskiy L, Sarrafzadegan N, Aznauryan A. Chemotherapy, hypothyroidism and oral dysbiosis as a novel risk factor of cardiovascular pathology development. Curr Probl Cardiol 2023; 48:101051. [PMID: 34800544 DOI: 10.1016/j.cpcardiol.2021.101051] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2021] [Revised: 11/04/2021] [Accepted: 11/05/2021] [Indexed: 02/01/2023]
Abstract
Cardiovascular disease (CVD) remains the leading cause of morbidity and mortality in the population, as well as the economic burden of the health care system. Currently, CVDs account for more than 17.6 million deaths a year and are projected to exceed 23.6 million by 2030. Unstable atheroma, and its rupture, underlies the pathology of most cardiovascular complications, particularly acute coronary syndrome, mortality from which, compared with other CV events, remains the leading one. Despite numerous efforts by WHO, national health systems, and medical authorities, the incidence and mortality from cardiovascular events remain critically high. Thus, the search for new risk factors for the development of CV pathology looks very relevant. Our working group decided to amalgamate our research data, which reflects the study of modern risk factors from the Armenian, Russian, Georgian, and Iranian medical schools. In particular, the aspects of cardiotoxic effects of chemotherapy, hypothyroidism, and oral dysbiosis are discussed.
Collapse
Affiliation(s)
- Ashot Avagimyan
- Lecturer of Anatomical Pathology Department, Yerevan State Medical University after M. Heratsi, Yerevan, Republic of Armenia
| | - Tamuna Gvianishvili
- Ivane Javakhishvili Tbilisi State University, Researcher of Department of Clinical and Experimental Pathology, Alexandre Natishvili Institute of Morphology, Ivane Javakhishvili Tbilisi State University, Tbilisi, Georgia
| | - Liana Gogiashvili
- Head of Department of Clinical and Experimental Pathology, Alexandre Natishvili Institute of Morphology, Ivane Javakhishvili Tbilisi State University, Tbilisi, Georgia
| | - Lev Kakturskiy
- Scientific Director of FSBI Research Institute of Human Morphology, President of Russian Society of Pathology, Moscow, Russia
| | - Nizal Sarrafzadegan
- Director of Isfahan Cardiovascular Research Institute, Isfahan University of Medical Science, Isfahan, Iran
| | - Artashes Aznauryan
- Histology Department, Yerevan State Medical University after M. Heratsi, Yerevan, Republic of Armenia
| |
Collapse
|
37
|
Implications of Inflammatory and Oxidative Stress Markers in the Attenuation of Nocturnal Blood Pressure Dipping. J Clin Med 2023; 12:jcm12041643. [PMID: 36836178 PMCID: PMC9959900 DOI: 10.3390/jcm12041643] [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: 12/22/2022] [Revised: 02/12/2023] [Accepted: 02/16/2023] [Indexed: 02/22/2023] Open
Abstract
To date, no model has jointly encompassed clinical, inflammatory, and redox markers with the risk of a non-dipper blood pressure (BP) profile. We aimed to evaluate the correlation between these features and the main twenty-four-hour ambulatory blood pressure monitoring (24-h ABPM) indices, as well as to establish a multivariate model including inflammatory, redox, and clinical markers for the prediction of a non-dipper BP profile. This was an observational study that included hypertensive patients older than 18 years. We enrolled 247 hypertensive patients (56% women) with a median age of 56 years. The results showed that higher levels of fibrinogen, tissue polypeptide-specific antigen, beta-2-microglobulin, thiobarbituric acid reactive substances, and copper/zinc ratio were associated with a higher risk of a non-dipper BP profile. Nocturnal systolic BP dipping showed a negative correlation with beta-globulin, beta-2-microglobulin, and gamma-globulin levels, whereas nocturnal diastolic BP dipping was positively correlated with alpha-2-globulin levels, and negatively correlated with gamma-globulin and copper levels. We found a correlation between nocturnal pulse pressure and beta-2-microglobulin and vitamin E levels, whereas the day-to-night pulse pressure gradient was correlated with zinc levels. Twenty-four-hour ABPM indices could exhibit singular inflammatory and redox patterns with implications that are still poorly understood. Some inflammatory and redox markers could be associated with the risk of a non-dipper BP profile.
Collapse
|
38
|
Effect of Peptides from Plasma of Patients with Coronary Artery Disease on the Vascular Endothelial Cells. MEDICINA (KAUNAS, LITHUANIA) 2023; 59:medicina59020238. [PMID: 36837440 PMCID: PMC10003965 DOI: 10.3390/medicina59020238] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Revised: 01/22/2023] [Accepted: 01/23/2023] [Indexed: 01/31/2023]
Abstract
Background and Objectives: Coronary artery disease (CAD) is the foremost cause of adult disability and mortality. There is an urgent need to focus on the research of new approaches for the prevention and treatment of CAD. Materials and Methods: The effects of peptides isolated from the blood plasma of CAD patients on endothelial cell secretion using the in vitro model have been tested. Human endothelial progenitor cells (HEPCs) were incubated for 24 h with peptides isolated from the plasma of healthy subjects or patients with stable angina, progressive unstable angina, and myocardial infarction. The contents of some soluble anticoagulant as well as procoagulant mediators in HEPC culture treated with peptide pools were then compared. Results and Conclusion: The results show that peptides from the plasma of patients with myocardial infarction promote endothelial cells to release both von Willebrand factor and endothelin-1, increasing vasoconstriction and shifting hemostatic balance toward a prothrombotic state. In contrast, peptides from the plasma of patients with progressive unstable angina suppress the secretion of endothelin-1 by HEPCs, while the secretion of both von Willebrand factor and tissue plasminogen activator was increased. As can be seen from the results obtained, disease derived peptides may contribute to the homeostasis of living organisms or the progression of pathological processes.
Collapse
|
39
|
Zhang W, Zhao J, Deng L, Ishimwe N, Pauli J, Wu W, Shan S, Kempf W, Ballantyne MD, Kim D, Lyu Q, Bennett M, Rodor J, Turner AW, Lu YW, Gao P, Choi M, Warthi G, Kim HW, Barroso MM, Bryant WB, Miller CL, Weintraub NL, Maegdefessel L, Miano JM, Baker AH, Long X. INKILN is a novel long noncoding RNA promoting vascular smooth muscle inflammation via scaffolding MKL1 and USP10. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.01.07.522948. [PMID: 36711681 PMCID: PMC9881896 DOI: 10.1101/2023.01.07.522948] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Background Activation of vascular smooth muscle cells (VSMCs) inflammation is vital to initiate vascular disease. However, the role of human-specific long noncoding RNAs (lncRNAs) in VSMC inflammation is poorly understood. Methods Bulk RNA-seq in differentiated human VSMCs revealed a novel human-specific lncRNA called IN flammatory M K L1 I nteracting L ong N oncoding RNA ( INKILN ). INKILN expression was assessed in multiple in vitro and ex vivo models of VSMC phenotypic modulation and human atherosclerosis and abdominal aortic aneurysm (AAA) samples. The transcriptional regulation of INKILN was determined through luciferase reporter system and chromatin immunoprecipitation assay. Both loss- and gain-of-function approaches and multiple RNA-protein and protein-protein interaction assays were utilized to uncover the role of INKILN in VSMC proinflammatory gene program and underlying mechanisms. Bacterial Artificial Chromosome (BAC) transgenic (Tg) mice were utilized to study INKLIN expression and function in ligation injury-induced neointimal formation. Results INKILN expression is downregulated in contractile VSMCs and induced by human atherosclerosis and abdominal aortic aneurysm. INKILN is transcriptionally activated by the p65 pathway, partially through a predicted NF-κB site within its proximal promoter. INKILN activates the proinflammatory gene expression in cultured human VSMCs and ex vivo cultured vessels. Mechanistically, INKILN physically interacts with and stabilizes MKL1, a key activator of VSMC inflammation through the p65/NF-κB pathway. INKILN depletion blocks ILIβ-induced nuclear localization of both p65 and MKL1. Knockdown of INKILN abolishes the physical interaction between p65 and MKL1, and the luciferase activity of an NF-κB reporter. Further, INKILN knockdown enhances MKL1 ubiquitination, likely through the reduced physical interaction with the deubiquitinating enzyme, USP10. INKILN is induced in injured carotid arteries and exacerbates ligation injury-induced neointimal formation in BAC Tg mice. Conclusions These findings elucidate an important pathway of VSMC inflammation involving an INKILN /MKL1/USP10 regulatory axis. Human BAC Tg mice offer a novel and physiologically relevant approach for investigating human-specific lncRNAs under vascular disease conditions.
Collapse
|
40
|
Fan J, Miao X, Zhang Z. Study on Maslinic Acid Inhibiting Cardiomyocyte Pyrolysis by Regulating TLR4/NF κB/NLRP3 Signaling Pathway. J BIOMATER TISS ENG 2023. [DOI: 10.1166/jbt.2023.3234] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Background: The paper aimed to investigate the effects of Maslinic acid on myocardial injury caused by ischemia and hypoxia after acute myocardial infarction, and to explore the mechanism of Maslinic acid inhibiting local inflammation and myocardial cell pyrolysis, slowing down
the synthesis of extracellular matrix, improving myocardial fibrosis and remodeling ventricle. Materials and Methods: Wistar rats with acute myocardial infarction were used. The serum levels of TNF-α, IL-1β, CRP, and LDH were detected by ELISA. The expressions
of inflammation-related proteins such as TLR4, NFκB, NLRP3, IL-10, and TGF-β in myocardial infarction were detected by Western-blot. TUNEL method was employed to detect myocardial cell pyrolysis. Masson staining was used to detect myocardial fibrosis. Immunohistochemical
method was applied to detect the expressions of CD206 and CD11C to understand the polarization of macrophages. Results: Maslinic acid can inhibit the levels of inflammatory factors such as CRP, TNF-α and IL-1β, and reduce the expression of myocardial TLR4, NFκB,
NLRP3 and other inflammation-related proteins after myocardial infarction. The release of LDH is significantly reduced. The expression of TUNEL positive cells in myocardium is obviously reduced after myocardial infarction. Maslinic acid can increase the expression of CD206-positive cells and
reduce the expression of CD11C-positive cells, thus regulate and promote polarization into M2 macrophages. Conclusion: Maslinic acid may inhibit myocardial cell pyrolysis and inflammatory response by regulating the TLR4/NFκB/NLRP3 signaling pathway. Maslinic acid can improve
myocardial fibrosis by inhibiting myocardial cell pyrolysis and regulating the polarization of macrophages.
Collapse
Affiliation(s)
- Jun Fan
- Department of Cardiology, Panyu Central Hospital, Guangzhou, 511400, China
| | - Xianghong Miao
- Department of Cardiology, Panyu Central Hospital, Guangzhou, 511400, China
| | - Zaiyong Zhang
- Department of Radiology, Panyu Central Hospital, Guangzhou, 511400, China
| |
Collapse
|
41
|
Di Francesco V, Di Francesco M, Palomba R, Brahmachari S, Decuzzi P, Ferreira M. Towards potent anti-inflammatory therapies in atherosclerosis: The case of methotrexate and colchicine combination into compartmentalized liposomes. J Drug Deliv Sci Technol 2023. [DOI: 10.1016/j.jddst.2023.104179] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
|
42
|
Tu S, He W, Han J, Wu A, Ren W. Advances in imaging and treatment of atherosclerosis based on organic nanoparticles. APL Bioeng 2022; 6:041501. [PMCID: PMC9726224 DOI: 10.1063/5.0127835] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2022] [Accepted: 10/31/2022] [Indexed: 12/09/2022] Open
Abstract
Atherosclerosis, a systemic chronic inflammatory disease, can lead to thrombosis and vascular occlusion, thereby inducing a series of serious vascular diseases. Currently, distinguishing unstable plaques early and achieving more effective treatment are the two main clinical concerns in atherosclerosis. Organic nanoparticles have great potential in atherosclerotic imaging and treatment, showing superior biocompatibility, drug-loading capacity, and synthesis. This article illustrates the process of atherosclerosis onset and the key targeted cells, then systematically summarizes recent progress made in organic nanoparticle-based imaging of different types of targeted cells and therapeutic methods for atherosclerosis, including optical and acoustic-induced therapy, drug delivery, gene therapy, and immunotherapy. Finally, we discuss the major impediments that need to be addressed in future clinical practice. We believe this article will help readers to develop a comprehensive and in-depth understanding of organic nanoparticle-based atherosclerotic imaging and treatment, thus advancing further development of anti-atherosclerosis therapies.
Collapse
Affiliation(s)
| | - Wenming He
- Department of Cardiology, The Affiliated Hospital of Medical School, Ningbo University, 247 Renmin Road, Jiangbei District, Ningbo, Zhejiang Province 315020, China,Authors to whom correspondence should be addressed:; ; and
| | | | - Aiguo Wu
- Authors to whom correspondence should be addressed:; ; and
| | - Wenzhi Ren
- Authors to whom correspondence should be addressed:; ; and
| |
Collapse
|
43
|
PCSK9 pathway-noncoding RNAs crosstalk: Emerging opportunities for novel therapeutic approaches in inflammatory atherosclerosis. Int Immunopharmacol 2022; 113:109318. [DOI: 10.1016/j.intimp.2022.109318] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Revised: 09/30/2022] [Accepted: 10/03/2022] [Indexed: 11/05/2022]
|
44
|
Lano G, Sallée M, Pelletier M, Bataille S, Fraisse M, McKay N, Brunet P, Dou L, Burtey S. Neutrophil:lymphocyte ratio correlates with the uremic toxin indoxyl sulfate and predicts the risk of death in patients on hemodialysis. Nephrol Dial Transplant 2022; 37:2528-2537. [PMID: 35146525 DOI: 10.1093/ndt/gfab350] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Chronic kidney disease (CKD) is a major public health issue associated with increased cardiovascular, infectious and all-cause mortality. The neutrophil:lymphocyte ratio (NLR) is a predictive marker of the risk of death and cardiovascular events. Uremic toxins, notably indoxyl sulfate (IS), are involved in immune deficiency and cardiovascular complications associated with CKD. The aim of this study was to assess whether the NLR was related to uremic toxins and could predict clinical outcome in hemodialysis (HD) patients. METHODS We conducted a prospective cohort study of 183 patients on chronic HD. The main objective was to study the correlation between the NLR and uremic toxin serum levels. The secondary objective was to test if the NLR can predict the incidence of mortality, cardiovascular events and infectious events. RESULTS Patients were separated into two groups according to the NLR median value (3.49). The NLR at inclusion was correlated with the NLR at the 6-month (r = 0.55, P < 0.0001) and 12-month (r = 0.62, P < 0.0001) follow-up. Among uremic toxins, IS levels were higher in the group with high NLR (104 µmol/L versus 81 µmol/L; P = 0.004). In multivariate analysis, the NLR remained correlated with IS (P = 0.03). The incidence of death, cardiovascular events and severe infectious events was higher in the group with high NLR [respectively, 38% versus 18% (P = 0.004), 45% versus 26% (P = 0.01) and 33% versus 21% (P = 0.02)] than in the low NLR group. Multivariate analysis showed an independent association of the NLR with mortality (P = 0.02) and cardiovascular events (P = 0.03) but not with severe infectious events. CONCLUSIONS In HD patients, the NLR predicted mortality and cardiovascular events but not severe infections and correlated positively with the level of the uremic toxin IS. The NLR could be an interesting marker for monitoring the risk of clinical events in CKD patients.
Collapse
Affiliation(s)
- Guillaume Lano
- Aix Marseille Université, INSERM, INRAE, C2VN, Marseille, France.,Centre de néphrologie et transplantation rénale, Hôpital de la conception AP-HM, Marseille, France
| | - Marion Sallée
- Aix Marseille Université, INSERM, INRAE, C2VN, Marseille, France.,Centre de néphrologie et transplantation rénale, Hôpital de la conception AP-HM, Marseille, France
| | - Marion Pelletier
- Centre de néphrologie et transplantation rénale, Hôpital de la conception AP-HM, Marseille, France
| | - Stanislas Bataille
- Aix Marseille Université, INSERM, INRAE, C2VN, Marseille, France.,Elsan, Phocean Institute of Nephrology, Clinique Bouchard, Marseille, France
| | - Megan Fraisse
- Aix Marseille Université, INSERM, INRAE, C2VN, Marseille, France
| | - Nathalie McKay
- Aix Marseille Université, INSERM, INRAE, C2VN, Marseille, France
| | - Philippe Brunet
- Aix Marseille Université, INSERM, INRAE, C2VN, Marseille, France.,Centre de néphrologie et transplantation rénale, Hôpital de la conception AP-HM, Marseille, France
| | - Laetitia Dou
- Aix Marseille Université, INSERM, INRAE, C2VN, Marseille, France
| | - Stéphane Burtey
- Aix Marseille Université, INSERM, INRAE, C2VN, Marseille, France.,Centre de néphrologie et transplantation rénale, Hôpital de la conception AP-HM, Marseille, France
| |
Collapse
|
45
|
Genetzakis E, Gilchrist J, Kassiou M, Figtree GA. Development and clinical translation of P2X7 receptor antagonists: A potential therapeutic target in coronary artery disease? Pharmacol Ther 2022; 237:108228. [DOI: 10.1016/j.pharmthera.2022.108228] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Revised: 05/17/2022] [Accepted: 06/06/2022] [Indexed: 12/12/2022]
|
46
|
Hadadi M, Mohseni-Badalabadi R, Hosseinsabet A. Effects of obesity on left atrial phasic functions in patients with chronic ischemic heart disease and preserved left ventricular ejection fraction without recent myocardial infarction: a two-dimensional speckle-tracking echocardiography study. J Ultrasound 2022; 25:521-527. [PMID: 34855185 PMCID: PMC9402816 DOI: 10.1007/s40477-021-00616-5] [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: 04/30/2021] [Accepted: 07/29/2021] [Indexed: 10/19/2022] Open
Abstract
PURPOSE Left atrial (LA) phasic functions in various subgroups of subjects with obesity are differently impaired, suggesting that obesity may have diverse effects in dissimilar subgroups of subjects with obesity. We aimed to compare the effects of obesity on LA phasic functions in patients suffering from chronic ischemic heart disease with a preserved left ventricular ejection fraction and without a recent myocardial infarction. METHODS In our cross-sectional study, 145 consecutive candidates for isolated coronary artery bypass graft (CABG) surgery were divided according to the presence of obesity into 2 groups: obese (36 patients) and nonobese (103 patients), and LA phasic functions were evaluated by two-dimensional speckle-tracking echocardiography (2D STE). RESULTS The longitudinal strain rate during the reservoir phase (3.0 ± 0.7 s-1 vs 2.7 ± 0.6 s-1; P = 0.032), longitudinal strain during the contraction phase (19.2 ± 4.7% vs 17.2 ± 4.1%; P = 0.022), and the longitudinal strain rate during the contraction phase (4.4 ± 1.2 s-1 vs 3.9 ± 1.2 s-1; P = 0.036) decreased in the obese group compared with the nonobese group. The other longitudinal 2D STE-derived markers of the LA phasic functions were not different between the 2 groups. CONCLUSIONS Among patients suffering from chronic ischemic heart disease with a preserved left ventricular ejection fraction and without a recent myocardial infarction, LA reservoir and contraction functions as measured by 2D STE were impaired in patients with obesity compared with those without it.
Collapse
Affiliation(s)
- Marjan Hadadi
- Cardiology Department, Tehran Heart Center, Tehran University of Medical Sciences, Karegar Shomali Street, Tehran, I.R. of Iran
| | - Reza Mohseni-Badalabadi
- Cardiology Department, Tehran Heart Center, Tehran University of Medical Sciences, Karegar Shomali Street, Tehran, I.R. of Iran
| | - Ali Hosseinsabet
- Cardiology Department, Tehran Heart Center, Tehran University of Medical Sciences, Karegar Shomali Street, Tehran, I.R. of Iran
| |
Collapse
|
47
|
Association Lp-PLA2 Gene Polymorphisms with Coronary Heart Disease. DISEASE MARKERS 2022; 2022:9775699. [PMID: 35818585 PMCID: PMC9271005 DOI: 10.1155/2022/9775699] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Accepted: 05/10/2022] [Indexed: 11/28/2022]
Abstract
Objectives The study evaluated the association between lipoprotein-associated phospholipase A2 (Lp-PLA2) gene polymorphisms and coronary heart disease (CHD), in order to explore the molecular genetics of CHD. Methods Groups of CHD patients (n = 283) and healthy controls (n = 261) were involved in this study. R92H, V279F, and A379V polymorphisms of LP-PLA2 gene were confirmed using polymerase chain reaction (PCR) and direct DNA sequencing. These polymorphisms and their interaction were also analyzed as potential risk factors of CHD. Results In this study population, the genotypes of R92H (GG, GA, and AA), V279F (CC, AC, and AA) and A379V (GG, GA, and AA) were studied. There was a significantly difference in frequencies of R92H between CHD patients and controls (P < 0.05). In contrast, no significant difference in frequencies of V279F and A379V existed between CHD patients and controls. Furthermore, R92H and A379V were in strong linkage disequilibrium. Conclusions These results suggested that R92H polymorphism might contribute to increased risk of CHD.
Collapse
|
48
|
Boczar KE, Shin S, Bezzina KA, Geejo A, Pearson AL, Shahab S, Fehlmann CA, Visintini S, Beanlands R, Wells GA. Examining anti-inflammatory therapies in the prevention of cardiovascular events: protocol for a systematic review and network meta-analysis of randomised controlled trials. BMJ Open 2022; 12:e062702. [PMID: 35760536 PMCID: PMC9237867 DOI: 10.1136/bmjopen-2022-062702] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
INTRODUCTION Inflammation is emerging as an important risk factor for atherosclerotic cardiovascular disease and has been a recent target for many novel therapeutic agents. However, comparative evidence regarding efficacy of these anti-inflammatory treatment options is currently lacking. METHODS AND ANALYSIS This systematic review will include randomised controlled trials evaluating the effect of anti-inflammatory agents on cardiovascular outcomes in patients with known cardiovascular disease. Studies will be retrieved from Medline, Embase, the Cochrane Central Register of Controlled Trials, as well as clinical trial registry websites, Europe PMC and conference abstract handsearching. No publication date or language restrictions will be imposed. Eligible interventions must have some component of anti-inflammatory agent. These include (but are not limited to): non-steroidal anti-inflammatory drugs (NSAIDs), colchicine, prednisone, methotrexate, canakinumab, pexelizumab, anakinra, succinobucol, losmapimod, inclacumab, atreleuton, LP-PLA2 (darapladib) and sPLA2 (varespladib). The primary outcomes will include major adverse cardiac events (MACE), and each individual component of MACE (myocardial infarction, stroke and cardiovascular death). Key secondary outcomes will include unstable angina, heart failure, all-cause mortality, cardiac arrest and revascularisation. Screening, inclusion, data extraction and quality assessment will be performed independently by two reviewers. Network meta-analysis based on the random effects model will be conducted to compare treatment effects both directly and indirectly. The quality of the evidence will be assessed with appropriate tools including the Grading of Recommendations, Assessment, Development and Evaluation profiler or Confidence in Network Meta-Analysis tool. ETHICS AND DISSEMINATION Ethics approval is not required for this systematic review. The findings will be disseminated through a peer-reviewed journal. PROSPERO REGISTRATION NUMBER CRD42022303289.
Collapse
Affiliation(s)
- Kevin Emery Boczar
- Cardiology, University of Ottawa Heart Institute, Ottawa, Ontario, Canada
- Epidemiology and Public Health, University of Ottawa Faculty of Medicine, Ottawa, Ontario, Canada
| | - Sheojung Shin
- Cardiology, University of Ottawa Heart Institute, Ottawa, Ontario, Canada
| | | | - Aishwarya Geejo
- Cardiology, University of Ottawa Heart Institute, Ottawa, Ontario, Canada
| | | | - Saba Shahab
- Department of Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | - Christophe A Fehlmann
- University of Ottawa, Ottawa, Ontario, Canada
- Geneva University Hospitals, Geneva, Switzerland
| | - Sarah Visintini
- Cardiology, University of Ottawa Heart Institute, Ottawa, Ontario, Canada
| | - Rob Beanlands
- Cardiology, University of Ottawa Heart Institute, Ottawa, Ontario, Canada
| | - George A Wells
- Cardiology, University of Ottawa Heart Institute, Ottawa, Ontario, Canada
- University of Ottawa, Ottawa, Ontario, Canada
| |
Collapse
|
49
|
Ramezani S, Rezaee SA, Farjami Z, Ebrahimi N, Abdullabass HK, Ibrahim Jebur MI, Rafatpanah H, Akbarin MM. HTLV, a multi organ oncovirus. Microb Pathog 2022; 169:105622. [DOI: 10.1016/j.micpath.2022.105622] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Revised: 06/03/2022] [Accepted: 06/06/2022] [Indexed: 12/29/2022]
|
50
|
Katanasaka Y, Saito A, Sunagawa Y, Sari N, Funamoto M, Shimizu S, Shimizu K, Akimoto T, Ueki C, Kitano M, Hasegawa K, Sakaguchi G, Morimoto T. ANGPTL4 Expression Is Increased in Epicardial Adipose Tissue of Patients with Coronary Artery Disease. J Clin Med 2022; 11:jcm11092449. [PMID: 35566578 PMCID: PMC9099928 DOI: 10.3390/jcm11092449] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Revised: 04/19/2022] [Accepted: 04/24/2022] [Indexed: 02/01/2023] Open
Abstract
Epicardial adipose tissue (EAT) is known to affect atherosclerosis and coronary artery disease (CAD) pathogenesis, persistently releasing pro-inflammatory adipokines that affect the myocardium and coronary arteries. Angiopoietin-like 4 (ANGPTL4) is a protein secreted from adipose tissue and plays a critical role in the progression of atherosclerosis. Here, the expression of ANGPTL4 in EAT was investigated in CAD subjects. Thirty-four consecutive patients (13 patients with significant CAD; 21 patients without CAD) undergoing elective open-heart surgery were recruited. EAT and pericardial fluid were obtained at the time of surgery. mRNA expression and ANGPTL4 and IL-1β levels were evaluated by qRT-PCR and ELISA. The expression of ANGPTL4 (p = 0.0180) and IL-1β (p < 0.0001) in EAT significantly increased in the CAD group compared to that in the non-CAD group and positively correlated (p = 0.004). Multiple regression analysis indicated that CAD is a contributing factor for ANGPTL4 expression in EAT. IL-1β level in the pericardial fluid was significantly increased in patients with CAD (p = 0.020). Moreover, the expression of ANGPTL4 (p = 0.004) and IL-1β (p < 0.001) in EAT was significantly increased in non-obese patients with CAD. In summary, ANGPTL4 expression in EAT was increased in CAD patients.
Collapse
Affiliation(s)
- Yasufumi Katanasaka
- Division of Molecular Medicine, School of Pharmaceutical Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka 422-8526, Japan; (Y.K.); (A.S.); (Y.S.); (N.S.); (M.F.); (S.S.); (K.S.); (K.H.)
- Division of Translational Research, National Hospital Organization Kyoto Medical Center, 1-1 Mukaihata-cho Fukakusa, Fushimi-ku, Kyoto 612-8555, Japan
- Laboratory of Clinical Cardiovascular Pharmacology, Shizuoka General Hospital, 4-27-1 Kita Ando Aoi-ku, Shizuoka 420-8527, Japan
| | - Ayumi Saito
- Division of Molecular Medicine, School of Pharmaceutical Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka 422-8526, Japan; (Y.K.); (A.S.); (Y.S.); (N.S.); (M.F.); (S.S.); (K.S.); (K.H.)
| | - Yoichi Sunagawa
- Division of Molecular Medicine, School of Pharmaceutical Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka 422-8526, Japan; (Y.K.); (A.S.); (Y.S.); (N.S.); (M.F.); (S.S.); (K.S.); (K.H.)
- Division of Translational Research, National Hospital Organization Kyoto Medical Center, 1-1 Mukaihata-cho Fukakusa, Fushimi-ku, Kyoto 612-8555, Japan
- Laboratory of Clinical Cardiovascular Pharmacology, Shizuoka General Hospital, 4-27-1 Kita Ando Aoi-ku, Shizuoka 420-8527, Japan
| | - Nurmila Sari
- Division of Molecular Medicine, School of Pharmaceutical Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka 422-8526, Japan; (Y.K.); (A.S.); (Y.S.); (N.S.); (M.F.); (S.S.); (K.S.); (K.H.)
| | - Masafumi Funamoto
- Division of Molecular Medicine, School of Pharmaceutical Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka 422-8526, Japan; (Y.K.); (A.S.); (Y.S.); (N.S.); (M.F.); (S.S.); (K.S.); (K.H.)
| | - Satoshi Shimizu
- Division of Molecular Medicine, School of Pharmaceutical Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka 422-8526, Japan; (Y.K.); (A.S.); (Y.S.); (N.S.); (M.F.); (S.S.); (K.S.); (K.H.)
| | - Kana Shimizu
- Division of Molecular Medicine, School of Pharmaceutical Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka 422-8526, Japan; (Y.K.); (A.S.); (Y.S.); (N.S.); (M.F.); (S.S.); (K.S.); (K.H.)
| | - Takehide Akimoto
- Department of Cardiovascular Surgery, Shizuoka General Hospital, 4-27-1 Kita Ando Aoi-ku, Shizuoka 420-8527, Japan; (T.A.); (C.U.); (M.K.)
| | - Chikara Ueki
- Department of Cardiovascular Surgery, Shizuoka General Hospital, 4-27-1 Kita Ando Aoi-ku, Shizuoka 420-8527, Japan; (T.A.); (C.U.); (M.K.)
| | - Mitsuru Kitano
- Department of Cardiovascular Surgery, Shizuoka General Hospital, 4-27-1 Kita Ando Aoi-ku, Shizuoka 420-8527, Japan; (T.A.); (C.U.); (M.K.)
| | - Koji Hasegawa
- Division of Molecular Medicine, School of Pharmaceutical Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka 422-8526, Japan; (Y.K.); (A.S.); (Y.S.); (N.S.); (M.F.); (S.S.); (K.S.); (K.H.)
- Division of Translational Research, National Hospital Organization Kyoto Medical Center, 1-1 Mukaihata-cho Fukakusa, Fushimi-ku, Kyoto 612-8555, Japan
| | - Genichi Sakaguchi
- Department of Cardiovascular Surgery, Kindai University Faculty of Medicine, 377-2 Ohno-Higashi, Osaka-Sayama 589-8511, Japan;
| | - Tatsuya Morimoto
- Division of Molecular Medicine, School of Pharmaceutical Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka 422-8526, Japan; (Y.K.); (A.S.); (Y.S.); (N.S.); (M.F.); (S.S.); (K.S.); (K.H.)
- Division of Translational Research, National Hospital Organization Kyoto Medical Center, 1-1 Mukaihata-cho Fukakusa, Fushimi-ku, Kyoto 612-8555, Japan
- Laboratory of Clinical Cardiovascular Pharmacology, Shizuoka General Hospital, 4-27-1 Kita Ando Aoi-ku, Shizuoka 420-8527, Japan
- Correspondence: ; Tel.: +81-54-264-5763
| |
Collapse
|