1
|
Li X, Lindmark B, Amilon C, Samuelsson K, Weidolf L, Nelander K, Knöchel J, Heijer M, Bragg RA, Gränfors M, Lindstedt E, Sidhu S, Garkaviy P, Ericsson H. Disposition of orally administered atuliflapon, a novel 5-lipoxygenase-activating protein inhibitor in healthy participants. Pharmacol Res Perspect 2024; 12:e70029. [PMID: 39400479 PMCID: PMC11472027 DOI: 10.1002/prp2.70029] [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: 03/21/2024] [Revised: 06/27/2024] [Accepted: 09/27/2024] [Indexed: 10/15/2024] Open
Abstract
In this study, the mass balance, pharmacokinetics (PK) and metabolism of atuliflapon, a novel 5-lipoxygenase-activating protein inhibitor, were investigated in healthy male subjects. A single oral dose of 200 mg [14C]atuliflapon suspension was administered to six healthy male subjects. Mass balance, PK and metabolite profiles of atuliflapon were analyzed using radioactivity monitoring and liquid chromatography with mass spectrometry analysis. The safety of atuliflapon was assessed during the study. Atuliflapon was rapidly absorbed with a median tmax of 1.5 h, followed by a biphasic decline in plasma exposure rendering a terminal half-life of ~20 h. Unchanged atuliflapon was the predominant radioactive component in plasma, accounting for 40.1% of the total drug-related exposure (DRE), while a direct N-glucuronide was the only metabolite exceeding 10% of DRE, accounting for 20.9%. Renal excretion of intact atuliflapon accounted for <1% of the administered dose. In total 85.2% of administered radioactivity was recovered over 312 h with 79.3% and 5.9% in feces and urine, respectively. Parent atuliflapon contributed to approximately 40% of the recovered dose in excreta, while metabolites resulting from phase 1 oxidative pathways accounted for more than 30% of the excreted dose. Overall, a single oral dose of 200 mg [14C]atuliflapon suspension was well tolerated in healthy male subjects. The human metabolism and disposition data obtained will support future development and submissions of atuliflapon as a potential candidate drug for the treatment of cardiovascular, cardiorenal, and respiratory indications.
Collapse
Affiliation(s)
- Xue‐Qing Li
- DMPK, Research and Early Development, CardiovascularRenal and Metabolism, BioPharmaceuticals R&D, AstraZenecaGothenburgSweden
| | - Bo Lindmark
- DMPK, Research and Early Development, CardiovascularRenal and Metabolism, BioPharmaceuticals R&D, AstraZenecaGothenburgSweden
| | - Carl Amilon
- DMPK, Research and Early Development, CardiovascularRenal and Metabolism, BioPharmaceuticals R&D, AstraZenecaGothenburgSweden
| | - Kristin Samuelsson
- DMPK, Research and Early Development, CardiovascularRenal and Metabolism, BioPharmaceuticals R&D, AstraZenecaGothenburgSweden
| | - Lars Weidolf
- DMPK, Research and Early Development, CardiovascularRenal and Metabolism, BioPharmaceuticals R&D, AstraZenecaGothenburgSweden
| | - Karin Nelander
- Biometrics CVRM, Late CVRMBioPharmaceuticals R&D, AstraZenecaGothenburgSweden
| | - Jane Knöchel
- Clinical Pharmacology and Quantitative Pharmacology, Clinical Pharmacology and Safety SciencesBioPharmaceuticals R&D, AstraZenecaGothenburgSweden
| | - Maria Heijer
- Integrated Bioanalysis, Clinical Pharmacology and Safety SciencesBioPharmaceuticals R&D, AstraZenecaGothenburgSweden
| | - Ryan A. Bragg
- Early Chemical DevelopmentPharmaceutical Sciences, R&D, AstraZenecaCambridgeUK
| | - Malin Gränfors
- Early Product Development and ManufacturingPharmaceutical Sciences, R&D, AstraZenecaGothenburgSweden
| | - Eva‐Lotte Lindstedt
- Projects, Research and Early Development, CardiovascularRenal and Metabolism, BioPharmaceuticals R&D, AstraZenecaGothenburgSweden
| | | | - Pavlo Garkaviy
- ECD, Research and Early Development, CardiovascularRenal and Metabolism, BioPharmaceuticals R&D, AstraZenecaGothenburgSweden
| | - Hans Ericsson
- Clinical Pharmacology and Quantitative Pharmacology, Clinical Pharmacology and Safety SciencesBioPharmaceuticals R&D, AstraZenecaGothenburgSweden
| |
Collapse
|
2
|
Kim J, Shin BS, Kim DH, Shin DI, Ahn SH, Kim JG, Ryu SH, Moon HR, Kang HG, Jeong H, Yum KS, Chae HY, Kim DH, Kang K, Kim J. Molecular genomic and epigenomic characteristics related to aspirin and clopidogrel resistance. BMC Med Genomics 2024; 17:166. [PMID: 38902747 PMCID: PMC11188263 DOI: 10.1186/s12920-024-01936-1] [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: 04/30/2024] [Accepted: 06/14/2024] [Indexed: 06/22/2024] Open
Abstract
BACKGROUND Mediators, genomic and epigenomic characteristics involving in metabolism of arachidonic acid by cyclooxygenase (COX) and lipoxygenase (ALOX) and hepatic activation of clopidogrel have been individually suggested as factors associated with resistance against aspirin and clopidogrel. The present multi-center prospective cohort study evaluated whether the mediators, genomic and epigenomic characteristics participating in arachidonic acid metabolism and clopidogrel activation could be factors that improve the prediction of the aspirin and clopidogrel resistance in addition to cardiovascular risks. METHODS We enrolled 988 patients with transient ischemic attack and ischemic stroke who were evaluated for a recurrence of ischemic stroke to confirm clinical resistance, and measured aspirin (ARU) and P2Y12 reaction units (PRU) using VerifyNow to assess laboratory resistance 12 weeks after aspirin and clopidogrel administration. We investigated whether mediators, genotypes, and promoter methylation of genes involved in COX and ALOX metabolisms and clopidogrel activation could synergistically improve the prediction of ischemic stroke recurrence and the ARU and PRU levels by integrating to the established cardiovascular risk factors. RESULTS The logistic model to predict the recurrence used thromboxane A synthase 1 (TXAS1, rs41708) A/A genotype and ALOX12 promoter methylation as independent variables, and, improved sensitivity of recurrence prediction from 3.4% before to 13.8% after adding the mediators, genomic and epigenomic variables to the cardiovascular risks. The linear model we used to predict the ARU level included leukotriene B4, COX2 (rs20417) C/G and thromboxane A2 receptor (rs1131882) A/A genotypes with the addition of COX1 and ALOX15 promoter methylations as variables. The linear PRU prediction model included G/A and prostaglandin I receptor (rs4987262) G/A genotypes, COX2 and TXAS1 promoter methylation, as well as cytochrome P450 2C19*2 (rs4244285) A/A, G/A, and *3 (rs4986893) A/A genotypes as variables. The linear models for predicting ARU (r = 0.291, R2 = 0.033, p < 0.01) and PRU (r = 0.503, R2 = 0.210, p < 0.001) levels had improved prediction performance after adding the genomic and epigenomic variables to the cardiovascular risks. CONCLUSIONS This study demonstrates that different mediators, genomic and epigenomic characteristics of arachidonic acid metabolism and clopidogrel activation synergistically improved the prediction of the aspirin and clopidogrel resistance together with the cardiovascular risk factors. TRIAL REGISTRATION URL: https://www. CLINICALTRIALS gov ; Unique identifier: NCT03823274.
Collapse
Grants
- YMC037 Yuhan Corporation, South Korea
- YMC037 Yuhan Corporation, South Korea
- YMC037 Yuhan Corporation, South Korea
- YMC037 Yuhan Corporation, South Korea
- YMC037 Yuhan Corporation, South Korea
- YMC037 Yuhan Corporation, South Korea
- YMC037 Yuhan Corporation, South Korea
- YMC037 Yuhan Corporation, South Korea
- YMC037 Yuhan Corporation, South Korea
- YMC037 Yuhan Corporation, South Korea
- YMC037 Yuhan Corporation, South Korea
Collapse
Affiliation(s)
- Jei Kim
- Department of Neurology, College of Medicine and Hospital, Daejeon-Chungnam Regional Cardiocerebrovascular Disease Center, Chungnam National University, Daejeon, South Korea.
- Department of Anatomy, College of Medicine, Chungnam National University, 266 Moonhwaro, Joongku, Daejeon, 35015, South Korea.
| | - Byoung-Soo Shin
- Department of Neurology, Research Institute of Clinical Medicine and Biomedical Research Institute, Medical School and Hospital, Jeonbuk National University, Jeonju, South Korea
| | - Dae-Hyun Kim
- Department of Neurology, Busan Regional Cardiocerebrovascular Disease Center, Dong-A University Hospital, Busan, South Korea
| | - Dong-Ick Shin
- Department of Neurology, Chungbuk Regional Cardiocerebrovascular Disease Center, Chungbuk National University Hospital, Cheongju, South Korea
| | - Seong Hwan Ahn
- Department of Neurology, Chosun University Hospital, Gwangju, South Korea
| | - Jae Guk Kim
- Department of Neurology, Eulji University Hospital, Daejeon, South Korea
| | - Su Hyun Ryu
- Department of Neurology, College of Medicine and Hospital, Daejeon-Chungnam Regional Cardiocerebrovascular Disease Center, Chungnam National University, Daejeon, South Korea
| | - Hye Rin Moon
- Department of Neurology, College of Medicine and Hospital, Daejeon-Chungnam Regional Cardiocerebrovascular Disease Center, Chungnam National University, Daejeon, South Korea
| | - Hyun Goo Kang
- Department of Neurology, Research Institute of Clinical Medicine and Biomedical Research Institute, Medical School and Hospital, Jeonbuk National University, Jeonju, South Korea
| | - Hyeseon Jeong
- Department of Neurology, College of Medicine and Hospital, Daejeon-Chungnam Regional Cardiocerebrovascular Disease Center, Chungnam National University, Daejeon, South Korea
| | - Kyu Sun Yum
- Department of Neurology, Chungbuk Regional Cardiocerebrovascular Disease Center, Chungbuk National University Hospital, Cheongju, South Korea
| | - Hee-Yun Chae
- Department of Neurology, Chungbuk Regional Cardiocerebrovascular Disease Center, Chungbuk National University Hospital, Cheongju, South Korea
| | - Do-Hyung Kim
- Department of Neurology, Eulji University Hospital, Daejeon, South Korea
| | - Keunsoo Kang
- Department of Microbiology, College of Science & Technology, Dankook University, Cheonan, South Korea
| | - Jeeyeon Kim
- Department of Neurology, College of Medicine and Hospital, Daejeon-Chungnam Regional Cardiocerebrovascular Disease Center, Chungnam National University, Daejeon, South Korea
| |
Collapse
|
3
|
Bi X, Wang Y, Lin Y, Wang M, Li X. Genetic Evidence for Causal Relationships between Plasma Eicosanoid Levels and Cardiovascular Disease. Metabolites 2024; 14:294. [PMID: 38921429 PMCID: PMC11206149 DOI: 10.3390/metabo14060294] [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: 04/17/2024] [Revised: 05/17/2024] [Accepted: 05/21/2024] [Indexed: 06/27/2024] Open
Abstract
Cardiovascular diseases are the most common causes of mortality and disability worldwide. Eicosanoids are a group of bioactive metabolites that are mainly oxidized by arachidonic acid. Eicosanoids play a diverse role in cardiovascular diseases, with some exerting beneficial effects while others have detrimental consequences. However, a causal relationship between eicosanoid levels and cardiovascular disease remains unclear. Six single nucleotide polymorphisms (SNPs) with strong associations with plasma eicosanoid levels were selected. Summary-level data for cardiovascular disease were obtained from publicly available genome-wide association studies. A two-sample MR analysis identified that plasma eicosanoid levels were inversely correlated with unstable angina pectoris (OR 1.06; 95% CI 1-1.12; p = 0.04), myocardial infarction (OR 1.05; 95% CI 1.02-1.09; p = 0.005), ischemia stroke (OR 1.05; 95% CI 1-1.11; p = 0.047), transient ischemic attack (OR 1.03; 95% CI 1-1.07; p = 0.042), heart failure (OR 1.03; 95% CI 1.01-1.05; p = 0.011), and pulmonary embolism (OR 1.08; 95% CI 1.02-1.14; p = 1.69 × 10-6). In conclusion, our data strongly suggest a genetic causal link between high plasma eicosanoid levels and an increased cardiovascular disease risk. This study provides genetic evidence for treating cardiovascular diseases.
Collapse
Affiliation(s)
- Xukun Bi
- Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang Province, Department of Cardiology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou 310016, China
| | - Yiran Wang
- Department of Nursing, No. 906 Hospital of People’s Liberation Army, Ningbo 315000, China
| | - Yangjun Lin
- Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang Province, Department of Cardiology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou 310016, China
| | - Meihui Wang
- Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang Province, Department of Cardiology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou 310016, China
| | - Xiaoting Li
- Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang Province, Department of Cardiology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou 310016, China
| |
Collapse
|
4
|
Ikram M, Shah I, Hussain H, Mughal EU, Naeem N, Sadiq A, Nazir Y, Ali Shah SW, Zahoor M, Ullah R, Ali EA, Umar MN. Synthesis, molecular docking evaluation for LOX and COX-2 inhibition and determination of in-vivo analgesic potentials of aurone derivatives. Heliyon 2024; 10:e29658. [PMID: 38694111 PMCID: PMC11058299 DOI: 10.1016/j.heliyon.2024.e29658] [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: 11/16/2023] [Revised: 04/11/2024] [Accepted: 04/11/2024] [Indexed: 05/03/2024] Open
Abstract
In the current study, seven (7) aurone derivatives (ADs) were synthesized and employed to in-vitro LOX and COX-2 assays, in-vivo models of acetic acid-induced mice writhing, formalin-induced mice paw licking and tail immersion test to evaluate their analgesic potential at the doses of 10 mg and 20 mg/kg body weight. Molecular docking was performed to know the active binding site at both LOX and COX-2 as compared to standard drugs. Among the ADs, 2-(3,4-dimethoxybenzylidene)benzofuran-3(2H)-one (WE-4)possessed optimal LOX and COX-2 inhibitory strength (IC50=0.30 μM and 0.22 μM) as compared to standard (ZileutonIC50 = 0.08 μM, CelecoxibIC50 = 0.05 μM). Similarly in various pain models compound WE-4 showed significantly (p < 0.05) highest percent analgesic potency as compared to control at a dose of 20 mg/kg i.e. 77.60 % analgesic effect in acetic acid model, 49.97 % (in Phase-1) and 70.93 % (inPhase-2) analgesic effect in formalin pain model and 74.71 % analgesic response in tail immersion model. By the administration of Naloxone, the tail flicking latencies were reversed (antagonized) in all treatments. The WE-4 (at 10 mg/kg and 20 mg/kg) was antagonized after 90 min from 11.23 ± 0.93 and 13.41 ± 1.21 to 5.30 ± 0.48 and 4.80 ± 0.61 respectively as compared to standard Tramadol (from 17.74 ± 1.33 to 3.70 ± 0.48), showing the opiodergic receptor involvement. The molecular docking study of ADs revealed that WE-4 had a higher affinity for LOX and COX-2 with docking scores of -4.324 and -5.843 respectively. As a whole, among the tested ADs, compound WE-4 demonstrated excellent analgesic effects that may have been caused by inhibiting the LOX and COX-2 pathways.
Collapse
Affiliation(s)
- Muhammad Ikram
- Department of Pharmacy, Abdul Wali Khan University Mardan (AWKUM), Mardan, 23390, Pakistan
| | - Ismail Shah
- Department of Pharmacy, Abdul Wali Khan University Mardan (AWKUM), Mardan, 23390, Pakistan
| | - Haya Hussain
- Department of Pharmacy, Shaheed Benazir Bhutto University Sheringal, Dir (Upper) 18000, Khyber Pakhtunkhwa, Pakistan
| | | | - Nafeesa Naeem
- Department of Chemistry, University of Gujrat, Gujrat, 50700, Pakistan
| | - Amina Sadiq
- Department of Chemistry, Govt. College Women University, Sialkot, 51300, Pakistan
| | - Yasir Nazir
- Department of Chemistry, University of Sialkot, Sialkot, 51300, Pakistan
| | - Syed Wadood Ali Shah
- Department of Pharmacy, University of Malakand, Chakdara, Chakdara 18800, Khyber Pakhtunkhwa, Pakistan
| | - Muhammad Zahoor
- Department of Biochemistry, University of Malakand, Chakdara, Dir Lower, KPK, 18800, Pakistan
| | - Riaz Ullah
- Department of Pharmacognosy, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Essam A. Ali
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | | |
Collapse
|
5
|
Tattersall MC, Jarjour NN, Busse PJ. Systemic Inflammation in Asthma: What Are the Risks and Impacts Outside the Airway? THE JOURNAL OF ALLERGY AND CLINICAL IMMUNOLOGY. IN PRACTICE 2024; 12:849-862. [PMID: 38355013 PMCID: PMC11219096 DOI: 10.1016/j.jaip.2024.02.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Revised: 01/19/2024] [Accepted: 02/02/2024] [Indexed: 02/16/2024]
Abstract
Airway inflammation in asthma has been well recognized for several decades, with general agreement on its role in asthma pathogenesis, symptoms, propensity toward exacerbation, and decline in lung function. This has led to universal recommendation in asthma management guidelines to incorporate the use of inhaled corticosteroid as an anti-inflammatory therapy for all patients with persistent asthma symptoms. However, there has been limited attention paid to the presence and potential impact of systemic inflammation in asthma. Accumulating evidence from epidemiological observations and cohort studies points to a host of downstream organ dysfunction in asthma especially among patients with longstanding or more severe disease, frequent exacerbations, and underlying risk factors for organ dysfunction. Most studies to date have focused on cognitive impairment, depression/anxiety, metabolic syndrome, and cardiovascular abnormalities. In this review, we summarize some of the evidence demonstrating these abnormalities and highlight the proposed mechanisms and potential benefits of treatment in limiting these extrapulmonary abnormalities in patients with asthma. The goal of this commentary is to raise awareness of the importance of recognizing potential extrapulmonary conditions associated with systemic inflammation of asthma. This area of treatment of patients with asthma is a large unmet need.
Collapse
Affiliation(s)
- Matthew C Tattersall
- Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, Wis.
| | - Nizar N Jarjour
- Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, Wis
| | - Paula J Busse
- Department of Medicine, Division of Clinical Immunology, Icahn School of Medicine at Mount Sinai School of Medicine, New York, NY
| |
Collapse
|
6
|
Kotlyarov S. Identification of Important Genes Associated with the Development of Atherosclerosis. Curr Gene Ther 2024; 24:29-45. [PMID: 36999180 DOI: 10.2174/1566523223666230330091241] [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: 09/17/2022] [Revised: 12/06/2022] [Accepted: 01/26/2023] [Indexed: 04/01/2023]
Abstract
Atherosclerosis is one of the most important medical problems due to its prevalence and significant contribution to the structure of temporary and permanent disability and mortality. Atherosclerosis is a complex chain of events occurring in the vascular wall over many years. Disorders of lipid metabolism, inflammation, and impaired hemodynamics are important mechanisms of atherogenesis. A growing body of evidence strengthens the understanding of the role of genetic and epigenetic factors in individual predisposition and development of atherosclerosis and its clinical outcomes. In addition, hemodynamic changes, lipid metabolism abnormalities, and inflammation are closely related and have many overlapping links in regulation. A better study of these mechanisms may improve the quality of diagnosis and management of such patients.
Collapse
Affiliation(s)
- Stanislav Kotlyarov
- Department of Nursing, Ryazan State Medical University Named After Academician I.P. Pavlov, Russian Federation
| |
Collapse
|
7
|
Wang X, Baskaran L, Chan M, Boisvert W, Hausenloy DJ. Targeting leukotriene biosynthesis to prevent atherosclerotic cardiovascular disease. CONDITIONING MEDICINE 2023; 6:33-41. [PMID: 38800614 PMCID: PMC11126214] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Indexed: 05/29/2024]
Abstract
Atherosclerotic cardiovascular disease (ASCVD) is the leading cause of death and disability worldwide. As such, new treatments are needed to prevent the onset and progression of atherosclerosis to improve outcomes in patients with coronary, cerebrovascular, and peripheral arterial disease. In this regard, inflammation is known to be a critical driver of atherosclerosis formation and progression, thus it is a viable target for vascular protection in patients at risk of developing ASCVD. Leukotrienes, key pro-inflammatory lipid mediators derived from arachidonic acid, are associated with atheroma inflammation and progression. Genetic mutations in key components of the leukotriene synthesis pathway, such as 5-lipoxygenase (5-LO) and 5-lipoxygenase-activating protein (FLAP), are associated with an increased risk of cardiovascular disease, and pharmacological inhibition of 5-LO and FLAP has been reported to prevent atheroma formation in pre-clinical and early clinical studies. In this article, we provide an overview of these studies and highlight the therapeutic potential of targeting leukotriene synthesis to prevent atheroma inflammation and progression and improve outcomes in patients at risk of ASCVD.
Collapse
Affiliation(s)
- Xiaomeng Wang
- Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore Medical School, Singapore
| | | | - Mark Chan
- Department of Cardiology, National University Heart Centre, National University Health System, Singapore
| | - William Boisvert
- Center for Cardiovascular Research, John A. Burns School of Medicine, University of Hawaii, USA
| | - Derek J Hausenloy
- Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore Medical School, Singapore
- National Heart Research Institute Singapore, National Heart Centre Singapore, Singapore
- Yong Loo Lin Medical School, National University of Singapore, Singapore
- The Hatter Cardiovascular Institute, University College London, London, UK
| |
Collapse
|
8
|
Guha Ray A, Odum OP, Wiseman D, Weinstock A. The diverse roles of macrophages in metabolic inflammation and its resolution. Front Cell Dev Biol 2023; 11:1147434. [PMID: 36994095 PMCID: PMC10041730 DOI: 10.3389/fcell.2023.1147434] [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: 01/18/2023] [Accepted: 02/14/2023] [Indexed: 03/14/2023] Open
Abstract
Macrophages are one of the most functionally diverse immune cells, indispensable to maintain tissue integrity and metabolic health. Macrophages perform a myriad of functions ranging from promoting inflammation, through inflammation resolution to restoring and maintaining tissue homeostasis. Metabolic diseases encompass a growing list of diseases which develop from a mix of genetics and environmental cues leading to metabolic dysregulation and subsequent inflammation. In this review, we summarize the contributions of macrophages to four metabolic conditions-insulin resistance and adipose tissue inflammation, atherosclerosis, non-alcoholic fatty liver disease and neurodegeneration. The role of macrophages is complex, yet they hold great promise as potential therapies to address these growing health concerns.
Collapse
Affiliation(s)
| | | | | | - Ada Weinstock
- Section of Genetic Medicine, Department of Medicine, The University of Chicago, Chicago, IL, United States
| |
Collapse
|
9
|
Eghtedari B, Roy SK, Budoff MJ. Anti-inflammatory Therapeutics and Coronary Artery Disease. Cardiol Rev 2023; 31:80-86. [PMID: 35471811 DOI: 10.1097/crd.0000000000000428] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
It has been demonstrated that atherosclerotic disease progression is contingent upon chronic inflammation. The sequence of events leading up to plaque formation, instability, and eventual plaque rupture hinges upon the interaction of proinflammatory cytokines and fat deposition within the coronary vasculature. Over the past decade, a large body of evidence has demonstrated the efficacy of specific anti-inflammatory therapeutics in halting the progression of coronary artery disease. Despite this, these therapeutics have yet to be included in guideline-directed medical therapy regimens. This review will focus on several anti-inflammatories, which have been studied in the context of cardiovascular disease-colchicine, canakinumab, VIA-2291, and methotrexate, and will highlight the potential benefits majority hold in hindering atherosclerosis and cardiovascular disease progression. This holds especially true for individuals already on optimal medical therapy who continue to be at high risk for adverse cardiovascular events.
Collapse
Affiliation(s)
- Bibinaz Eghtedari
- From the The Lundquist Institute, Harbor-University of California-Los Angeles, Torrance, CA
| | | | | |
Collapse
|
10
|
Dodd KE, Blackley DJ, Mazurek JM. Cardiovascular Disease Among Adults With Work-Related Asthma, 2012-2017. Am J Prev Med 2023; 64:194-203. [PMID: 36371324 DOI: 10.1016/j.amepre.2022.09.010] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Revised: 08/23/2022] [Accepted: 09/12/2022] [Indexed: 11/11/2022]
Abstract
INTRODUCTION Asthma is associated with an increased risk for cardiovascular disease, and adults with persistent, severe asthma have a significantly higher risk of cardiovascular disease than adults with intermittent or no asthma. METHODS The objective of this cross-sectional study was to assess the association between work-related asthma status and cardiovascular disease among ever-employed adults (aged 18-64 years) with current asthma using data from the 2012-2017 Behavioral Risk Factor Surveillance System Asthma Call-Back Survey from 37 states and the District of Columbia. Weighted prevalence ratios and 95% CIs, adjusted for age, sex, race/ethnicity, education, household income, smoking status, chronic obstructive pulmonary disease, diabetes, and BMI, were calculated. In addition, the associations of cardiovascular disease with adverse asthma outcomes and asthma control among adults with work-related asthma were examined. Analyses were conducted in 2021. RESULTS Among an estimated annualized 14.8 million ever-employed adults aged 18-64 years with current asthma, adults with work-related asthma (prevalence ratio=1.5; 95% CI=1.2, 1.8) and possible work-related asthma (prevalence ratio=1.2; 95% CI=1.0, 1.5) were significantly more likely to have cardiovascular disease than adults with non-work-related asthma. Among adults with work-related asthma, those with very poorly controlled asthma (prevalence ratio=1.8; 95% CI=1.3, 2.5) and an asthma-related emergency room visit (prevalence ratio=1.5; 95% CI=1.1, 2.0) were significantly more likely to have cardiovascular disease. CONCLUSIONS Adults with work-related asthma were more likely to have cardiovascular disease than those with non-work-related asthma. Primary prevention, early diagnosis, and implementation of optimal work-related asthma management are essential for workers' health. Cardiovascular disease should be considered where appropriate when diagnosing and recommending treatment and interventions for adults with work-related asthma.
Collapse
Affiliation(s)
- Katelynn E Dodd
- Respiratory Health Division, National Institute for Occupational Safety and Health, Centers for Disease Control and Prevention (CDC), Morgantown, West Virginia.
| | - David J Blackley
- Respiratory Health Division, National Institute for Occupational Safety and Health, Centers for Disease Control and Prevention (CDC), Morgantown, West Virginia
| | - Jacek M Mazurek
- Respiratory Health Division, National Institute for Occupational Safety and Health, Centers for Disease Control and Prevention (CDC), Morgantown, West Virginia
| |
Collapse
|
11
|
Tattersall MC. Asthma as a Systemic Disease: Cardiovascular Effects Associated with Asthma. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2023; 1426:77-100. [PMID: 37464117 DOI: 10.1007/978-3-031-32259-4_4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/20/2023]
Abstract
Asthma and cardiovascular disease (CVD) pose significant public health burdens. Airway inflammation is central to asthma pathophysiology and systemic inflammation, which occurs in asthma, is central to CVD pathophysiology. Numerous robust epidemiological studies have demonstrated deleterious systemic cardiovascular effects associated with the asthma syndrome. The cardiovascular effects associated with asthma include arterial injury, atherosclerotic CVD events, atrial fibrillation, and hypertension. Asthma is a heterogeneous disease, however, and the risk of CVD is not homogeneous across the various clinical phenotypes and molecular endotypes, highlighting prior inconsistent associations of asthma and its subtypes with various forms of CVD. The mechanistic underpinnings of the increased CVD risk in asthma remain multifactorial and undefined. Collectively, this supports the need for a precision approach in the identification of individuals with asthma who remain at elevated risk of development of cardiovascular diseases to guide both diagnostic and preventive interventions to decrease CVD risk among individuals living with asthma.
Collapse
|
12
|
Blockade of the BLT1-LTB 4 axis does not affect mast cell migration towards advanced atherosclerotic lesions in LDLr -/- mice. Sci Rep 2022; 12:18362. [PMID: 36319730 PMCID: PMC9626554 DOI: 10.1038/s41598-022-23162-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Accepted: 10/26/2022] [Indexed: 11/05/2022] Open
Abstract
Mast cells have been associated with the progression and destabilization of advanced atherosclerotic plaques. Reducing intraplaque mast cell accumulation upon atherosclerosis progression could be a potent therapeutic strategy to limit plaque destabilization. Leukotriene B4 (LTB4) has been reported to induce mast cell chemotaxis in vitro. Here, we examined whether antagonism of the LTB4-receptor BLT1 could inhibit mast cell accumulation in advanced atherosclerosis. Expression of genes involved in LTB4 biosynthesis was determined by single-cell RNA sequencing of human atherosclerotic plaques. Subsequently, Western-type diet fed LDLr-/- mice with pre-existing atherosclerosis were treated with the BLT1-antagonist CP105,696 or vehicle control three times per week by oral gavage. In the spleen, a significant reduction in CD11b+ myeloid cells was observed, including Ly6Clo and Ly6Chi monocytes as well as dendritic cells. However, atherosclerotic plaque size, collagen and macrophage content in the aortic root remained unaltered upon treatment. Finally, BLT1 antagonism did not affect mast cell numbers in the aortic root. Here, we show that human intraplaque leukocytes may be a source of locally produced LTB4. However, BLT1-antagonism during atherosclerosis progression does not affect either local mast cell accumulation or plaque size, suggesting that other mechanisms participate in mast cell accumulation during atherosclerosis progression.
Collapse
|
13
|
Kotlyarov S. Genetic and Epigenetic Regulation of Lipoxygenase Pathways and Reverse Cholesterol Transport in Atherogenesis. Genes (Basel) 2022; 13:1474. [PMID: 36011386 PMCID: PMC9408222 DOI: 10.3390/genes13081474] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Revised: 08/12/2022] [Accepted: 08/16/2022] [Indexed: 11/16/2022] Open
Abstract
Atherosclerosis is one of the most important medical and social problems of modern society. Atherosclerosis causes a large number of hospitalizations, disability, and mortality. A considerable amount of evidence suggests that inflammation is one of the key links in the pathogenesis of atherosclerosis. Inflammation in the vascular wall has extensive cross-linkages with lipid metabolism, and lipid mediators act as a central link in the regulation of inflammation in the vascular wall. Data on the role of genetics and epigenetic factors in the development of atherosclerosis are of great interest. A growing body of evidence is strengthening the understanding of the significance of gene polymorphism, as well as gene expression dysregulation involved in cross-links between lipid metabolism and the innate immune system. A better understanding of the genetic basis and molecular mechanisms of disease pathogenesis is an important step towards solving the problems of its early diagnosis and treatment.
Collapse
Affiliation(s)
- Stanislav Kotlyarov
- Department of Nursing, Ryazan State Medical University, 390026 Ryazan, Russia
| |
Collapse
|
14
|
Recent advances in function and structure of two leukotriene B 4 receptors: BLT1 and BLT2. Biochem Pharmacol 2022; 203:115178. [PMID: 35850310 DOI: 10.1016/j.bcp.2022.115178] [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: 05/27/2022] [Revised: 07/12/2022] [Accepted: 07/12/2022] [Indexed: 11/21/2022]
Abstract
Leukotriene B4 (LTB4) is generated by the enzymatic oxidation of arachidonic acid, which is then released from the cell membrane and acts as a potent activator of leukocytes and other inflammatory cells. Numerous studies have demonstrated the physiological and pathophysiological significance of this lipid in various diseases. LTB4 exerts its activities by binding to its specific G protein-coupled receptors (GPCRs): BLT1 and BLT2. In mouse disease models, treatment with BLT1 antagonists or BLT1 gene ablation attenuated various diseases, including bronchial asthma, arthritis, and psoriasis, whereas BLT2 deficiency exacerbated several diseases in the skin, cornea, and small intestine. Therefore, BLT1 inhibitors and BLT2 activators could be beneficial for the treatment of several inflammatory and immune disorders. As a result, attractive compounds targeting LTB4 receptors have been developed by several pharmaceutical companies. This review aims to understand the potential of BLT1 and BLT2 as therapeutic targets for the treatment of various inflammatory diseases. In addition, recent topics are discussed with major focuses on the structure and post-translational modifications of BLT1 and BLT2. Collectively, current evidence on modulating LTB4 receptor functions provides new strategies for the treatment of various diseases.
Collapse
|
15
|
Prescott E, Angerås O, Erlinge D, Grove EL, Hedman M, Jensen LO, Pernow J, Saraste A, Åkerblom A, Svedlund S, Rudvik A, Knöchel J, Lindstedt EL, Garkaviy P, Gan LM, Gabrielsen A. Safety and efficacy of the 5-lipoxygenase-activating protein inhibitor AZD5718 in patients with recent myocardial infarction: The phase 2a FLAVOUR study. Int J Cardiol 2022; 365:34-40. [PMID: 35842004 DOI: 10.1016/j.ijcard.2022.07.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Accepted: 07/12/2022] [Indexed: 11/05/2022]
Abstract
BACKGROUND Leukotrienes are pro-inflammatory vasoactive lipid mediators implicated in the pathophysiology of atherosclerotic cardiovascular disease. We studied the effect of the 5-lipoxygenase-activating protein inhibitor AZD5718 on leukotriene biosynthesis and coronary microvascular function in a single-blind, phase 2a study. METHODS Patients 7-28 days after myocardial infarction (±ST elevation), with <50% left anterior descending coronary artery stenosis and Thrombolysis in Myocardial Infarction flow grade ≥ 2 after percutaneous coronary intervention, were randomized 2:1:2 to once-daily AZD5718 200 mg or 50 mg, or placebo, in 4- and 12-week cohorts. Change in urine leukotriene E4 (uLTE4) was the primary endpoint, and coronary flow velocity reserve (CFVR; via echocardiography) was the key secondary endpoint. RESULTS Of 129 randomized patients, 128 received treatment (200 mg, n = 52; 50 mg, n = 25; placebo, n = 51). Statistically significant reductions in uLTE4 levels of >80% were observed in both AZD5718 groups versus the placebo group at 4 and 12 weeks. No significant changes in CFVR were observed for AZD5718 versus placebo. Adverse events (AEs) occurred in 12/18, 3/6 and 6/13 patients receiving 200 mg, 50 mg and placebo, respectively, in the 4-week cohort, and in 27/34, 14/19 and 24/38 patients, respectively, in the 12-week cohort. Serious AEs in seven patients receiving AZD5718 and four receiving placebo were not treatment-related, and there were no deaths. CONCLUSIONS In patients with recent myocardial infarction, AZD5718 was well tolerated, and leukotriene biosynthesis was dose-dependently inhibited. No significant changes in CFVR were detected. CLINICALTRIALS gov identifier: NCT03317002.
Collapse
Affiliation(s)
- Eva Prescott
- Department of Cardiology, Bispebjerg Hospital, University of Copenhagen, Copenhagen, Denmark.
| | - Oskar Angerås
- Department of Molecular and Clinical Medicine, Sahlgrenska Academy, University of Gothenburg, and Department of Cardiology, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - David Erlinge
- Cardiology, Department of Clinical Sciences, Lund University and Skåne University Hospital, Lund, Sweden
| | - Erik L Grove
- Department of Cardiology, Aarhus University Hospital, Aarhus, Denmark; Department of Clinical Medicine, Faculty of Health, Aarhus University, Aarhus, Denmark
| | - Marja Hedman
- Heart Center and Clinical Imaging Center, Kuopio University Hospital, Kuopio, Finland; Institute of Medicine, University of Eastern Finland, Kuopio, Finland
| | - Lisette O Jensen
- Department of Cardiology, Odense University Hospital, Odense, Denmark
| | - John Pernow
- Division of Cardiology, Department of Medicine, Karolinska Institute, and Department of Cardiology, Karolinska University Hospital, Stockholm, Sweden
| | - Antti Saraste
- University of Turku and Heart Centre, Turku University Hospital, Turku, Finland
| | - Axel Åkerblom
- Department of Medical Sciences - Cardiology, and Uppsala Clinical Research Center, Uppsala University, Uppsala, Sweden
| | - Sara Svedlund
- Department of Clinical Physiology, Sahlgrenska University Hospital and Department of Molecular and Clinical Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Anna Rudvik
- Early Biometrics and Statistical Innovation, Data Science & AI, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Jane Knöchel
- Clinical Pharmacology and Quantitative Pharmacology, Clinical Pharmacology and Safety Sciences, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Eva-Lotte Lindstedt
- Research and Early Clinical Development, Cardiovascular, Renal and Metabolic, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Pavlo Garkaviy
- Research and Early Clinical Development, Cardiovascular, Renal and Metabolic, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Li-Ming Gan
- Department of Molecular and Clinical Medicine, Sahlgrenska Academy, University of Gothenburg, and Department of Cardiology, Sahlgrenska University Hospital, Gothenburg, Sweden; Research and Early Clinical Development, Cardiovascular, Renal and Metabolic, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Anders Gabrielsen
- Research and Early Clinical Development, Cardiovascular, Renal and Metabolic, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| |
Collapse
|
16
|
McGurk KA, Farrell L, Kendall AC, Keavney BD, Nicolaou A. Genetic analyses of circulating PUFA-derived mediators identifies heritable dihydroxyeicosatrienoic acid species. Prostaglandins Other Lipid Mediat 2022; 160:106638. [PMID: 35472599 DOI: 10.1016/j.prostaglandins.2022.106638] [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: 12/13/2021] [Revised: 03/30/2022] [Accepted: 04/20/2022] [Indexed: 10/18/2022]
Abstract
Estimates of heritability are the first step in identifying a trait with substantial variation due to genetic factors. Large-scale genetic analyses can identify the DNA variants that influence the levels of circulating lipid species and the statistical technique Mendelian randomisation can use these DNA variants to address potential causality of these lipids in disease. We estimated the heritability of plasma eicosanoids, octadecanoids and docosanoids to identify those lipid species with substantial heritability. We analysed plasma lipid mediators in 31 White British families (196 participants) ascertained for high blood pressure and deeply clinically and biochemically phenotyped over a 25-year period. We found that the dihydroxyeicosatrienoic acid (DHET) species, 11,12-DHET and 14,15-DHET, products of arachidonic acid metabolism by cytochrome P450 (CYP) monooxygenase and soluble epoxide hydrolase (sEH), exhibited substantial heritability (h2 = 33%-37%; Padj<0.05). Identification of these two heritable bioactive lipid species allows for future large-scale, targeted, lipidomics-genomics analyses to address causality in cardiovascular and other diseases.
Collapse
Affiliation(s)
- Kathryn A McGurk
- Division of Cardiovascular Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, UK; Laboratory for Lipidomics and Lipid Research, Division of Pharmacy and Optometry, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, UK
| | - Laura Farrell
- Laboratory for Lipidomics and Lipid Research, Division of Pharmacy and Optometry, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, UK
| | - Alexandra C Kendall
- Laboratory for Lipidomics and Lipid Research, Division of Pharmacy and Optometry, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, UK
| | - Bernard D Keavney
- Manchester Heart Centre, Manchester University NHS Foundation Trust, UK
| | - Anna Nicolaou
- Laboratory for Lipidomics and Lipid Research, Division of Pharmacy and Optometry, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, UK.
| |
Collapse
|
17
|
Evans BR, Yerly A, van der Vorst EPC, Baumgartner I, Bernhard SM, Schindewolf M, Döring Y. Inflammatory Mediators in Atherosclerotic Vascular Remodeling. Front Cardiovasc Med 2022; 9:868934. [PMID: 35600479 PMCID: PMC9114307 DOI: 10.3389/fcvm.2022.868934] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Accepted: 04/11/2022] [Indexed: 12/23/2022] Open
Abstract
Atherosclerotic vascular disease remains the most common cause of ischemia, myocardial infarction, and stroke. Vascular function is determined by structural and functional properties of the arterial vessel wall, which consists of three layers, namely the adventitia, media, and intima. Key cells in shaping the vascular wall architecture and warranting proper vessel function are vascular smooth muscle cells in the arterial media and endothelial cells lining the intima. Pathological alterations of this vessel wall architecture called vascular remodeling can lead to insufficient vascular function and subsequent ischemia and organ damage. One major pathomechanism driving this detrimental vascular remodeling is atherosclerosis, which is initiated by endothelial dysfunction allowing the accumulation of intimal lipids and leukocytes. Inflammatory mediators such as cytokines, chemokines, and modified lipids further drive vascular remodeling ultimately leading to thrombus formation and/or vessel occlusion which can cause major cardiovascular events. Although it is clear that vascular wall remodeling is an elementary mechanism of atherosclerotic vascular disease, the diverse underlying pathomechanisms and its consequences are still insufficiently understood.
Collapse
Affiliation(s)
- Bryce R. Evans
- Division of Angiology, Swiss Cardiovascular Center, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
- Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland
| | - Anaïs Yerly
- Division of Angiology, Swiss Cardiovascular Center, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
- Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland
| | - Emiel P. C. van der Vorst
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians-University Munich (LMU), Munich, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Munich Heart Alliance, Munich, Germany
- Institute for Molecular Cardiovascular Research (IMCAR) and Interdisciplinary Center for Clinical Research (IZKF), RWTH Aachen University, Aachen, Germany
- Department of Pathology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Centre, Maastricht, Netherlands
| | - Iris Baumgartner
- Division of Angiology, Swiss Cardiovascular Center, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
- Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland
| | - Sarah Maike Bernhard
- Division of Angiology, Swiss Cardiovascular Center, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
- Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland
| | - Marc Schindewolf
- Division of Angiology, Swiss Cardiovascular Center, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
- Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland
| | - Yvonne Döring
- Division of Angiology, Swiss Cardiovascular Center, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
- Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians-University Munich (LMU), Munich, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Munich Heart Alliance, Munich, Germany
- *Correspondence: Yvonne Döring
| |
Collapse
|
18
|
Manubolu VS, Budoff MJ. Achieving coronary plaque regression: a decades-long battle against coronary artery disease. Expert Rev Cardiovasc Ther 2022; 20:291-305. [PMID: 35466832 DOI: 10.1080/14779072.2022.2069559] [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] [Indexed: 10/18/2022]
Abstract
INTRODUCTION Traditionally atherosclerosis was thought to be progressive and medical treatment solely focused on delaying the progression of atherosclerosis rather than treating the disease itself. Multiple recent studies, however, have demonstrated a significant decrease in cardiovascular mortality with the use of additional anti-atherosclerotic therapies beyond statins. Consistent with these observations, mechanistic studies indicate that these additional anti-atherosclerotic therapies have a positive effect on both halting and reversing the course of atherosclerosis. AREAS COVERED We examine the progression of atherosclerosis and the efficacy of various anti-atherosclerotic treatment classes in this review utilizing multimodality imaging techniques. Searches were conducted in electronic databases: PubMed and EMBASE for all peer reviewed publications that examined coronary plaque progression, regression and stabilization using different imaging modalities and antiatherosclerosis therapies. The keywords coronary plaque, coronary angiography, IVUS, intravascular OCT, CCTA in conjunction with the various therapies included in this review were searched in different combinations. All relevant published articles on this topic were identified and their reference lists were screened for relevance. EXPERT COMMENTARY Though lipoprotein levels have traditionally been the target for antiatherosclerosis medication, several newer strategies have emerged creating novel targets in the treatment of coronary atherosclerosis. Using a combination of antiatherosclerosis therapies in conjunction with noninvasive imaging modalities like CCTA to directly visualize the plaque, is currently the focus of the future, with the aim of preventing and reversing atherosclerosis.
Collapse
Affiliation(s)
| | - Matthew J Budoff
- Department of Cardiology, Lundquist Institute, Torrance, CA, USA
| |
Collapse
|
19
|
Grigorieva NY, Ilushina TP, Kolosova KS. The possibilities of using beta-blocker bisoprolol in patients with stable angina with concomitant bronchial asthma. KARDIOLOGIIA 2022; 62:32-39. [PMID: 35168531 DOI: 10.18087/cardio.2022.1.n1714] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Accepted: 10/29/2021] [Indexed: 06/14/2023]
Abstract
Aim To compare efficacy and safety of treatments with the calcium antagonist (CA) verapamil, the cardioselective β-blocker (BB) bisoprolol, and a combination therapy with bisoprolol and amlodipine in patients with stable angina (SA) with concurrent mild and moderate, persistent bronchial asthma (BA). Material and methods This open, prospective, randomized, comparative study included 120 patients with an IHD+BA comorbidity. Of these patients, 60 had mild persistent BA and 60 had moderate persistent BA. Each group was divided into 3 subgroup, each including 20 patients, based on the used regimen of antianginal therapy. Stepwise dose titration was performed every 2 weeks (subgroup 1 received the BB bisoprolol 2.5 mg - 5 mg - 10 mg; subgroup 2 received the CA verapamil 240 mg - 240 mg - 240 mg; subgroup 3 received bisoprolol 2.5 mg followed by the combination treatment with bisoprolol and amlodipine as a fixed combination 5+5 mg). All patients underwent a complete clinical and instrumental examination at baseline and at 2, 4, and 6 weeks of treatment. The antianginal effectivity and the effect on bronchial patency were evaluated. Results In patients with SA and mild persistent BA, the study of external respiration function (ERF) at 2, 4, and 6 weeks of treatment did not detect any significant difference in the forced expiratory volume in 1 second (FEV1) between the treatment subgroups. In patients with SA and moderate persistent BA receiving the treatment, a significant decrease in FEV1 (р=0.022) was observed in subgroup 1 receiving bisoprolol 10 mg at 6 weeks of treatment. In subgroups 2 and 3 during the treatment, significant differences were absent. In patients with SA and mild or moderate persistent BA, the heart rate was significantly decreased in all three subgroups; however, in subgroup 2 receiving verapamil, the changes were considerably smaller than in other subgroups.Conclusion The study results showed that the BB bisoprolol with dose titration every two weeks from 2.5 to 10 mg or the combination treatment with the BB bisoprolol and the CA amlodipine can be used as the antianginal therapy in patients with SA and mild persistent BA. The BB bisoprolol may be used in patients with SA and moderate persistent BA as the antianginal therapy, but only at doses not exceeding 5 mg to avoid the development of bronchial obstruction. The combination therapy with the BB bisoprolol 5 mg and the CA amlodipine 5 mg is indicated to enhance antianginal and vasoprotective effects.
Collapse
Affiliation(s)
- N Yu Grigorieva
- National Research N.I. Lobachevsky State University of Nizhny Novgorod, Nizhny Novgorod, Russia
| | - T P Ilushina
- "Central City Hospital of Arzamas", Arzamas, Russia
| | - K S Kolosova
- National Research N.I. Lobachevsky State University of Nizhny Novgorod, Nizhny Novgorod, Russia
| |
Collapse
|
20
|
Kotlyarov S, Kotlyarova A. Involvement of Fatty Acids and Their Metabolites in the Development of Inflammation in Atherosclerosis. Int J Mol Sci 2022; 23:1308. [PMID: 35163232 PMCID: PMC8835729 DOI: 10.3390/ijms23031308] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Revised: 01/14/2022] [Accepted: 01/21/2022] [Indexed: 02/06/2023] Open
Abstract
Despite all the advances of modern medicine, atherosclerosis continues to be one of the most important medical and social problems. Atherosclerosis is the cause of several cardiovascular diseases, which are associated with high rates of disability and mortality. The development of atherosclerosis is associated with the accumulation of lipids in the arterial intima and the disruption of mechanisms that maintain the balance between the development and resolution of inflammation. Fatty acids are involved in many mechanisms of inflammation development and maintenance. Endothelial cells demonstrate multiple cross-linkages between lipid metabolism and innate immunity. In addition, these processes are linked to hemodynamics and the function of other cells in the vascular wall, highlighting the central role of the endothelium in vascular biology.
Collapse
Affiliation(s)
- Stanislav Kotlyarov
- Department of Nursing, Ryazan State Medical University, 390026 Ryazan, Russia
| | - Anna Kotlyarova
- Department of Pharmacology and Pharmacy, Ryazan State Medical University, 390026 Ryazan, Russia;
| |
Collapse
|
21
|
Zhou Y, Liang ZS, Jin Y, Ding J, Huang T, Moore JH, Zheng ZJ, Huang J. Shared Genetic Architecture and Causal Relationship Between Asthma and Cardiovascular Diseases: A Large-Scale Cross-Trait Analysis. Front Genet 2022; 12:775591. [PMID: 35126453 PMCID: PMC8811262 DOI: 10.3389/fgene.2021.775591] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Accepted: 12/08/2021] [Indexed: 12/11/2022] Open
Abstract
Background: Accumulating evidence has suggested that there is a positive association between asthma and cardiovascular diseases (CVDs), implying a common architecture between them. However, the shared genetic architecture and causality of asthma and CVDs remain unclear. Methods: Based on the genome-wide association study (GWAS) summary statistics of recently published studies, our study examined the genetic correlation, shared genetic variants, and causal relationship between asthma (N = 127,669) and CVDs (N = 86,995–521,612). Statistical methods included high-definition likelihood (HDL), cross-trait meta-analyses of large-scale GWAS, transcriptome-wide association studies (TWAS), and Mendelian randomization (MR). Results: First, we observed a significant genetic correlation between asthma and heart failure (HF) (Rg = 0.278, P = 5 × 10−4). Through cross-trait analyses, we identified a total of 145 shared loci between asthma and HF. Fifteen novel loci were not previously reported for association with either asthma or HF. Second, we mapped these 145 loci to a total of 99 genes whose expressions are enriched in a broad spectrum of tissues, including the seminal vesicle, tonsil, appendix, spleen, skin, lymph nodes, breast, cervix and uterus, skeletal muscle, small intestine, lung, prostate, cardiac muscle, and liver. TWAS analysis identified five significant genes shared between asthma and HF in tissues from the hemic and immune system, digestive system, integumentary system, and nervous system. GSDMA, GSDMB, and ORMDL3 are statistically independent genetic effects from all shared TWAS genes between asthma and HF. Third, through MR analysis, genetic liability to asthma was significantly associated with heart failure at the Bonferroni-corrected significance level. The odds ratio (OR) is 1.07 [95% confidence interval (CI): 1.03–1.12; p = 1.31 × 10−3] per one-unit increase in loge odds of asthma. Conclusion: These findings provide strong evidence of genetic correlations and causal relationship between asthma and HF, suggesting a shared genetic architecture for these two diseases.
Collapse
Affiliation(s)
- Yi Zhou
- Department of Global Health, School of Public Health, Peking University, Beijing, China
| | - Zhi-Sheng Liang
- Department of Global Health, School of Public Health, Peking University, Beijing, China
| | - Yinzi Jin
- Department of Global Health, School of Public Health, Peking University, Beijing, China
| | - Jiayuan Ding
- College of Arts and Sciences, Boston University, Boston, MA, United States
| | - Tao Huang
- Department of Global Health, School of Public Health, Peking University, Beijing, China
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University, Beijing, China
| | - Jason H. Moore
- Department of Biostatistics, Epidemiology and Informatics, Institute for Biomedical Informatics, University of Pennsylvania, Philadelphia, PA, United States
| | - Zhi-Jie Zheng
- Department of Global Health, School of Public Health, Peking University, Beijing, China
- Institute for Global Health and Development, Peking University, Beijing, China
| | - Jie Huang
- Department of Global Health, School of Public Health, Peking University, Beijing, China
- Institute for Global Health and Development, Peking University, Beijing, China
- *Correspondence: Jie Huang,
| |
Collapse
|
22
|
Gerges SH, El-Kadi AOS. Sex differences in eicosanoid formation and metabolism: A possible mediator of sex discrepancies in cardiovascular diseases. Pharmacol Ther 2021; 234:108046. [PMID: 34808133 DOI: 10.1016/j.pharmthera.2021.108046] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Revised: 11/07/2021] [Accepted: 11/16/2021] [Indexed: 12/14/2022]
Abstract
Arachidonic acid is metabolized by cyclooxygenase, lipoxygenase, and cytochrome P450 enzymes to produce prostaglandins, leukotrienes, epoxyeicosatrienoic acids (EETs), and hydroxyeicosatetraenoic acids (HETEs), along with other eicosanoids. Eicosanoids have important physiological and pathological roles in the body, including the cardiovascular system. Evidence from several experimental and clinical studies indicates differences in eicosanoid levels, as well as in the activity or expression levels of their synthesizing and metabolizing enzymes between males and females. In addition, there is a clear state of gender specificity in cardiovascular diseases (CVD), which tend to be more common in men compared to women, and their risk increases significantly in postmenopausal women compared to younger women. This could be largely attributed to sex hormones, as androgens exert detrimental effects on the heart and blood vessels, whereas estrogen exhibits cardioprotective effects. Many of androgen and estrogen effects on the cardiovascular system are mediated by eicosanoids. For example, androgens increase the levels of cardiotoxic eicosanoids like 20-HETE, while estrogens increase the levels of cardioprotective EETs. Thus, sex differences in eicosanoid levels in the cardiovascular system could be an important underlying mechanism for the different effects of sex hormones and the differences in CVD between males and females. Understanding the role of eicosanoids in these differences can help improve the management of CVD.
Collapse
Affiliation(s)
- Samar H Gerges
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, Alberta, Canada
| | - Ayman O S El-Kadi
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, Alberta, Canada.
| |
Collapse
|
23
|
Pokhrel S, Gudneppanavar R, Teegala LR, Duah E, Thodeti CK, Paruchuri S. Leukotriene D 4 Upregulates Oxidized Low-Density Lipoprotein Receptor 1 and CD36 to Enhance Oxidized LDL Uptake and Phagocytosis in Macrophages Through Cysteinyl Leukotriene Receptor 1. Front Physiol 2021; 12:756450. [PMID: 34867460 PMCID: PMC8637273 DOI: 10.3389/fphys.2021.756450] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Accepted: 10/14/2021] [Indexed: 11/20/2022] Open
Abstract
Endothelial permeability, leukocyte attachment, and unregulated oxidized LDL (oxLDL) uptake by macrophages leading to the formation of foam cells are all vital in the initiation and progression of atherosclerosis. During inflammation, several inflammatory mediators regulate this process through the expression of distinct oxLDL binding cell surface receptors on macrophages. We have previously shown that Leukotriene D4 (LTD4) promotes endothelial dysfunction, increasing endothelial permeability and enhancing TNFα-mediated attachment of monocytes to endothelium, which hints at its possible role in atherosclerosis. Here we analyzed the effect of LTD4 on macrophage function. Macrophages mainly express CysLT1R and flux calcium in response to LTD4. Further, LTD4 potentiates phagocytosis in macrophages as revealed by the uptake of zymosan particles. Notably, LTD4 augmented macrophage phagocytosis and oxLDL uptake which is sensitive to MK-571 [Montelukast (MK)], a CysLT1R-specific antagonist. Mechanistically, LTD4 upregulated two receptors central to foam cell formation, oxidized low-density lipoprotein receptor-1 (OLR1/LOX-1), and CD36 in a time and dose-dependent manner. Finally, LTD4 enhanced the secretion of chemokines MCP-1 and MIP1β. Our results suggest that LTD4 contributes to atherosclerosis either through driving foam cell formation or recruitment of immune cells or both. CysLT1R antagonists are safely being used in the treatment of asthma, and the findings from the current study suggest that these can be re-purposed for the treatment of atherosclerosis.
Collapse
Affiliation(s)
- Sabita Pokhrel
- Department of Chemistry, University of Akron, Akron, OH, United States
| | | | - Lakshminarayan Reddy Teegala
- Department of Chemistry, University of Akron, Akron, OH, United States
- Department of Physiology and Pharmacology, University of Toledo College of Medicine and Life Sciences, Toledo, OH, United States
| | - Ernest Duah
- Department of Chemistry, University of Akron, Akron, OH, United States
| | - Charles K. Thodeti
- Department of Physiology and Pharmacology, University of Toledo College of Medicine and Life Sciences, Toledo, OH, United States
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown, OH, United States
| | - Sailaja Paruchuri
- Department of Chemistry, University of Akron, Akron, OH, United States
- Department of Physiology and Pharmacology, University of Toledo College of Medicine and Life Sciences, Toledo, OH, United States
| |
Collapse
|
24
|
Liu M, Yan M, He J, Lv H, Chen Z, Peng L, Cai W, Yao F, Chen C, Shi L, Zhang K, Zhang X, Wang DW, Wang L, Zhu Y, Ai D. Macrophage MST1/2 Disruption Impairs Post-Infarction Cardiac Repair via LTB4. Circ Res 2021; 129:909-926. [PMID: 34515499 DOI: 10.1161/circresaha.121.319687] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
[Figure: see text].
Collapse
MESH Headings
- Animals
- Antigens, CD/genetics
- Antigens, CD/metabolism
- Antigens, Differentiation, Myelomonocytic/genetics
- Antigens, Differentiation, Myelomonocytic/metabolism
- Chemokine CCL2/genetics
- Chemokine CCL2/metabolism
- Chemokine CCL4/genetics
- Chemokine CCL4/metabolism
- Chemokine CXCL2/metabolism
- Female
- Leukotriene B4/metabolism
- Lipoxygenase/metabolism
- Macrophages/metabolism
- Male
- Mice
- Mice, Inbred C57BL
- Myocardial Infarction/metabolism
- Myocardium/metabolism
- Protein Serine-Threonine Kinases/genetics
- Protein Serine-Threonine Kinases/metabolism
- Receptors, Cell Surface/genetics
- Receptors, Cell Surface/metabolism
- Receptors, Leukotriene B4/antagonists & inhibitors
- Receptors, Leukotriene B4/metabolism
- Serine-Threonine Kinase 3/genetics
- Serine-Threonine Kinase 3/metabolism
Collapse
Affiliation(s)
- Mingming Liu
- Tianjin Key Laboratory of Ion and Molecular Function of Cardiovascular Diseases, Tianjin Institute of Cardiology, State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), the Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics (M.L., M.Y., H.L., D.A.), Tianjin Medical University
- Neurology, Tianjin Neurological Institute, Tianjin Medical University General Hospital (M.L.)
| | - Meng Yan
- Tianjin Key Laboratory of Ion and Molecular Function of Cardiovascular Diseases, Tianjin Institute of Cardiology, State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), the Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics (M.L., M.Y., H.L., D.A.), Tianjin Medical University
- The First Affiliated Hospital of Soochow University Department of Pathology, Soochow University, Suzhou (M.Y.)
| | - Jinlong He
- Physiology and Pathophysiology (J.H., H.L., Z.C., W.C., X.Z., Y.Z., D.A.), Tianjin Medical University
| | - Huizhen Lv
- Tianjin Key Laboratory of Ion and Molecular Function of Cardiovascular Diseases, Tianjin Institute of Cardiology, State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), the Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics (M.L., M.Y., H.L., D.A.), Tianjin Medical University
- Physiology and Pathophysiology (J.H., H.L., Z.C., W.C., X.Z., Y.Z., D.A.), Tianjin Medical University
| | - Zhipeng Chen
- Physiology and Pathophysiology (J.H., H.L., Z.C., W.C., X.Z., Y.Z., D.A.), Tianjin Medical University
| | - Liyuan Peng
- Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science & Technology, Wuhan (L.P., C.C., D.-W.W.)
| | - Wenbin Cai
- Physiology and Pathophysiology (J.H., H.L., Z.C., W.C., X.Z., Y.Z., D.A.), Tianjin Medical University
| | - Fang Yao
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College (F.Y., L.W.)
| | - Chen Chen
- Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science & Technology, Wuhan (L.P., C.C., D.-W.W.)
| | - Lei Shi
- Biochemistry and Molecular Biology, School of Basic Medical Sciences (L.S., K.Z.), Tianjin Medical University
| | - Kai Zhang
- Biochemistry and Molecular Biology, School of Basic Medical Sciences (L.S., K.Z.), Tianjin Medical University
| | - Xu Zhang
- Physiology and Pathophysiology (J.H., H.L., Z.C., W.C., X.Z., Y.Z., D.A.), Tianjin Medical University
| | - Dao-Wen Wang
- Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science & Technology, Wuhan (L.P., C.C., D.-W.W.)
| | - Li Wang
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College (F.Y., L.W.)
| | - Yi Zhu
- Physiology and Pathophysiology (J.H., H.L., Z.C., W.C., X.Z., Y.Z., D.A.), Tianjin Medical University
| | - Ding Ai
- Tianjin Key Laboratory of Ion and Molecular Function of Cardiovascular Diseases, Tianjin Institute of Cardiology, State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), the Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics (M.L., M.Y., H.L., D.A.), Tianjin Medical University
- Physiology and Pathophysiology (J.H., H.L., Z.C., W.C., X.Z., Y.Z., D.A.), Tianjin Medical University
| |
Collapse
|
25
|
Han YH, Lee K, Saha A, Han J, Choi H, Noh M, Lee YH, Lee MO. Specialized Proresolving Mediators for Therapeutic Interventions Targeting Metabolic and Inflammatory Disorders. Biomol Ther (Seoul) 2021; 29:455-464. [PMID: 34162770 PMCID: PMC8411019 DOI: 10.4062/biomolther.2021.094] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Revised: 06/07/2021] [Accepted: 06/08/2021] [Indexed: 12/18/2022] Open
Abstract
Uncontrolled inflammation is considered the pathophysiological basis of many prevalent metabolic disorders, such as nonalcoholic fatty liver disease, diabetes, obesity, and neurodegenerative diseases. The inflammatory response is a self-limiting process that produces a superfamily of chemical mediators, called specialized proresolving mediators (SPMs). SPMs include the ω-3-derived family of molecules, such as resolvins, protectins, and maresins, as well as arachidonic acid-derived (ω-6) lipoxins that stimulate and promote resolution of inflammation, clearance of microbes, and alleviation of pain and promote tissue regeneration via novel mechanisms. SPMs function by binding and activating G protein-coupled receptors, such as FPR2/ALX, GPR32, and ERV1, and nuclear orphan receptors, such as RORα. Recently, several studies reported that SPMs have the potential to attenuate lipid metabolism disorders. However, the understanding of pharmacological aspects of SPMs, including tissue-specific biosynthesis, and specific SPM receptors and signaling pathways, is currently limited. Here, we summarize recent advances in the role of SPMs in resolution of inflammatory diseases with metabolic disorders, such as nonalcoholic fatty liver disease and obesity, obtained from preclinical animal studies. In addition, the known SPM receptors and their intracellular signaling are reviewed as targets of resolution of inflammation, and the currently available information on the therapeutic effects of major SPMs for metabolic disorders is summarized.
Collapse
Affiliation(s)
- Yong-Hyun Han
- Laboratory of Pathology and Physiology, College of Pharmacy, Kangwon National University, Chuncheon 24341, Republic of Korea
| | - Kyeongjin Lee
- Laboratory of Pathology and Physiology, College of Pharmacy, Kangwon National University, Chuncheon 24341, Republic of Korea
| | - Abhirup Saha
- College of Pharmacy, Seoul National University, Seoul 08826, Republic of Korea
| | - Juhyeong Han
- College of Pharmacy, Seoul National University, Seoul 08826, Republic of Korea
| | - Haena Choi
- College of Pharmacy, Seoul National University, Seoul 08826, Republic of Korea
| | - Minsoo Noh
- College of Pharmacy, Seoul National University, Seoul 08826, Republic of Korea
| | - Yun-Hee Lee
- College of Pharmacy, Seoul National University, Seoul 08826, Republic of Korea
| | - Mi-Ock Lee
- College of Pharmacy, Seoul National University, Seoul 08826, Republic of Korea.,Bio-MAX Institute, Seoul National University, Seoul 08826, Republic of Korea
| |
Collapse
|
26
|
Giménez-Bastida JA, González-Sarrías A, Laparra-Llopis JM, Schneider C, Espín JC. Targeting Mammalian 5-Lipoxygenase by Dietary Phenolics as an Anti-Inflammatory Mechanism: A Systematic Review. Int J Mol Sci 2021; 22:7937. [PMID: 34360703 PMCID: PMC8348464 DOI: 10.3390/ijms22157937] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Revised: 07/20/2021] [Accepted: 07/21/2021] [Indexed: 12/15/2022] Open
Abstract
5-Lipoxygenase (5-LOX) plays a key role in inflammation through the biosynthesis of leukotrienes and other lipid mediators. Current evidence suggests that dietary (poly)phenols exert a beneficial impact on human health through anti-inflammatory activities. Their mechanisms of action have mostly been associated with the modulation of pro-inflammatory cytokines (TNF-α, IL-1β), prostaglandins (PGE2), and the interaction with NF-κB and cyclooxygenase 2 (COX-2) pathways. Much less is known about the 5-lipoxygenase (5-LOX) pathway as a target of dietary (poly)phenols. This systematic review aimed to summarize how dietary (poly)phenols target the 5-LOX pathway in preclinical and human studies. The number of studies identified is low (5, 24, and 127 human, animal, and cellular studies, respectively) compared to the thousands of studies focusing on the COX-2 pathway. Some (poly)phenolics such as caffeic acid, hydroxytyrosol, resveratrol, curcumin, nordihydroguaiaretic acid (NDGA), and quercetin have been reported to reduce the formation of 5-LOX eicosanoids in vitro. However, the in vivo evidence is inconclusive because of the low number of studies and the difficulty of attributing effects to (poly)phenols. Therefore, increasing the number of studies targeting the 5-LOX pathway would largely expand our knowledge on the anti-inflammatory mechanisms of (poly)phenols.
Collapse
Affiliation(s)
- Juan Antonio Giménez-Bastida
- Laboratory of Food and Health, Research Group on Quality, Safety and Bioactivity of Plant Foods, Department Food Science and Technology, CEBAS-CSIC, P.O. Box 164, Campus de Espinardo, 30100 Murcia, Spain;
| | - Antonio González-Sarrías
- Laboratory of Food and Health, Research Group on Quality, Safety and Bioactivity of Plant Foods, Department Food Science and Technology, CEBAS-CSIC, P.O. Box 164, Campus de Espinardo, 30100 Murcia, Spain;
| | - José Moisés Laparra-Llopis
- Group of Molecular Immunonutrition in Cancer, Madrid Institute for Advanced Studies in Food (IMDEA-Food), 28049 Madrid, Spain;
| | - Claus Schneider
- Division of Clinical Pharmacology, Department of Pharmacology, Vanderbilt Institute of Chemical Biology, Vanderbilt University Medical School, Nashville, TN 37232, USA;
| | - Juan Carlos Espín
- Laboratory of Food and Health, Research Group on Quality, Safety and Bioactivity of Plant Foods, Department Food Science and Technology, CEBAS-CSIC, P.O. Box 164, Campus de Espinardo, 30100 Murcia, Spain;
| |
Collapse
|
27
|
Niksirat H, Siino V, Steinbach C, Levander F. High-Resolution Proteomic Profiling Shows Sexual Dimorphism in Zebrafish Heart-Associated Proteins. J Proteome Res 2021; 20:4075-4088. [PMID: 34185526 DOI: 10.1021/acs.jproteome.1c00387] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Understanding the molecular basis of sexual dimorphism in the cardiovascular system may contribute to the improvement of the outcome in biological, pharmacological, and toxicological studies as well as on the development of sex-based drugs and therapeutic approaches. Label-free protein quantification using high-resolution mass spectrometry was applied to detect sex-based proteome differences in the heart of zebrafish Danio rerio. Out of almost 3000 unique identified proteins in the heart, 79 showed significant abundance differences between male and female fish. The functional differences were mapped using enrichment analyses. Our results suggest that a large amount of materials needed for reproduction (e.g., sugars, lipids, proteins, etc.) may impose extra pressure on blood, vessels, and heart on their way toward the ovaries. In the present study, the female's heart shows a clear sexual dimorphism by changing abundance levels of numerous proteins, which could be a way to safely overcome material-induced elevated pressures. These proteins belong to the immune system, oxidative stress response, drug metabolization, detoxification, energy, metabolism, and so on. In conclusion, we showed that sex can induce dimorphism at the molecular level in nonsexual organs such as heart and must be considered as an important factor in cardiovascular research. Data are available via ProteomeXchange with identifier PXD023506.
Collapse
Affiliation(s)
- Hamid Niksirat
- Faculty of Fisheries and Protection of Waters, CENAKVA, University of South Bohemia in České Budějovice, Vodňany, 370 05 České Budějovice, Czech Republic
| | - Valentina Siino
- Department of Immunotechnology, Lund University, Lund 223 87, Sweden
| | - Christoph Steinbach
- Faculty of Fisheries and Protection of Waters, CENAKVA, University of South Bohemia in České Budějovice, Vodňany, 370 05 České Budějovice, Czech Republic
| | - Fredrik Levander
- Department of Immunotechnology, Lund University, Lund 223 87, Sweden.,National Bioinformatics Infrastructure Sweden (NBIS), Science for Life Laboratory, Lund University, Lund 223 87, Sweden
| |
Collapse
|
28
|
Wang B, Wu L, Chen J, Dong L, Chen C, Wen Z, Hu J, Fleming I, Wang DW. Metabolism pathways of arachidonic acids: mechanisms and potential therapeutic targets. Signal Transduct Target Ther 2021; 6:94. [PMID: 33637672 PMCID: PMC7910446 DOI: 10.1038/s41392-020-00443-w] [Citation(s) in RCA: 616] [Impact Index Per Article: 154.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Revised: 10/04/2020] [Accepted: 10/15/2020] [Indexed: 01/31/2023] Open
Abstract
The arachidonic acid (AA) pathway plays a key role in cardiovascular biology, carcinogenesis, and many inflammatory diseases, such as asthma, arthritis, etc. Esterified AA on the inner surface of the cell membrane is hydrolyzed to its free form by phospholipase A2 (PLA2), which is in turn further metabolized by cyclooxygenases (COXs) and lipoxygenases (LOXs) and cytochrome P450 (CYP) enzymes to a spectrum of bioactive mediators that includes prostanoids, leukotrienes (LTs), epoxyeicosatrienoic acids (EETs), dihydroxyeicosatetraenoic acid (diHETEs), eicosatetraenoic acids (ETEs), and lipoxins (LXs). Many of the latter mediators are considered to be novel preventive and therapeutic targets for cardiovascular diseases (CVD), cancers, and inflammatory diseases. This review sets out to summarize the physiological and pathophysiological importance of the AA metabolizing pathways and outline the molecular mechanisms underlying the actions of AA related to its three main metabolic pathways in CVD and cancer progression will provide valuable insight for developing new therapeutic drugs for CVD and anti-cancer agents such as inhibitors of EETs or 2J2. Thus, we herein present a synopsis of AA metabolism in human health, cardiovascular and cancer biology, and the signaling pathways involved in these processes. To explore the role of the AA metabolism and potential therapies, we also introduce the current newly clinical studies targeting AA metabolisms in the different disease conditions.
Collapse
Affiliation(s)
- Bei Wang
- Division of Cardiology, Department of Internal Medicine and Gene Therapy Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Huazhong University of Science and Technology, Hubei Province, Wuhan, China
- Department of Rheumatology and Immunology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Hubei, Wuhan, China
| | - Lujin Wu
- Division of Cardiology, Department of Internal Medicine and Gene Therapy Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Huazhong University of Science and Technology, Hubei Province, Wuhan, China
| | - Jing Chen
- Division of Cardiology, Department of Internal Medicine and Gene Therapy Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Huazhong University of Science and Technology, Hubei Province, Wuhan, China
| | - Lingli Dong
- Department of Rheumatology and Immunology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Hubei, Wuhan, China
| | - Chen Chen
- Division of Cardiology, Department of Internal Medicine and Gene Therapy Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Huazhong University of Science and Technology, Hubei Province, Wuhan, China
| | - Zheng Wen
- Division of Cardiology, Department of Internal Medicine and Gene Therapy Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Huazhong University of Science and Technology, Hubei Province, Wuhan, China
| | - Jiong Hu
- Institute for Vascular Signalling, Centre for Molecular Medicine, Goethe University, Frankfurt am Main, Germany
| | - Ingrid Fleming
- Institute for Vascular Signalling, Centre for Molecular Medicine, Goethe University, Frankfurt am Main, Germany
| | - Dao Wen Wang
- Division of Cardiology, Department of Internal Medicine and Gene Therapy Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Huazhong University of Science and Technology, Hubei Province, Wuhan, China.
| |
Collapse
|
29
|
Sabotin RP, Varon A, Roa JA, Raghuram A, Ishii D, Nino M, Galloy AE, Patel D, Raghavan ML, Hasan D, Samaniego EA. Insights into the pathogenesis of cerebral fusiform aneurysms: high-resolution MRI and computational analysis. J Neurointerv Surg 2021; 13:1180-1186. [PMID: 33632878 DOI: 10.1136/neurintsurg-2020-017243] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Revised: 01/26/2021] [Accepted: 01/29/2021] [Indexed: 11/04/2022]
Abstract
BACKGROUND Intracranial fusiform aneurysms are complex and poorly characterized vascular lesions. High-resolution magnetic resonance imaging (HR-MRI) and computational morphological analysis may be used to characterize cerebral fusiform aneurysms. OBJECTIVE To use advanced imaging and computational analysis to understand the unique pathophysiology, and determine possible underlying mechanisms of instability of cerebral fusiform aneurysms. METHODS Patients with unruptured intracranial aneurysms prospectively underwent imaging with 3T HR-MRI at diagnosis. Aneurysmal wall enhancement was objectively quantified using signal intensity after normalization of the contrast ratio (CR) with the pituitary stalk. Enhancement between saccular and fusiform aneurysms was compared, as well as enhancement characteristics of fusiform aneurysms. The presence of microhemorrhages in fusiform aneurysms was determined with quantitative susceptibility mapping (QSM). Three distinct types of fusiform aneurysms were analyzed with computational fluid dynamics (CFD) and finite element analysis (FEA). RESULTS A total of 130 patients with 160 aneurysms underwent HR-MRI. 136 aneurysms were saccular and 24 were fusiform. Fusiform aneurysms had a significantly higher CR and diameter than saccular aneurysms. Enhancing fusiform aneurysms exhibited more enhancement of reference vessels than non-enhancing fusiform aneurysms. Ten fusiform aneurysms underwent QSM analysis, and five aneurysms showed microhemorrhages. Microhemorrhage-positive aneurysms had a larger volume, diameter, and greater enhancement than aneurysms without microhemorrhage. Three types of fusiform aneurysms exhibited different CFD and FEA patterns. CONCLUSION Fusiform aneurysms exhibited more contrast enhancement than saccular aneurysms. Enhancing fusiform aneurysms had larger volume and diameter, more enhancement of reference vessels, and more often exhibited microhemorrhage than non-enhancing aneurysms. CFD and FEA suggest that various pathophysiological processes determine the formation and growth of fusiform aneurysms.
Collapse
Affiliation(s)
- Ryan Phillip Sabotin
- Department of Neurology, The University of Iowa Hospitals and Clinics, Iowa City, Iowa, USA
| | - Alberto Varon
- Department of Neurology, The University of Iowa, Iowa City, Iowa, USA
| | - Jorge A Roa
- Department of Neurology, The University of Iowa Hospitals and Clinics, Iowa City, Iowa, USA
| | - Ashrita Raghuram
- Department of Neurology, The University of Iowa Hospitals and Clinics, Iowa City, Iowa, USA
| | - Daizo Ishii
- Department of Neurosurgery, The University of Iowa Hospitals and Clinics, Iowa City, Iowa, USA
| | - Marco Nino
- Roy J Carver Department of Biomedical Engineering, The University of Iowa, Iowa City, Iowa, USA
| | - Adam E Galloy
- Roy J Carver Department of Biomedical Engineering, The University of Iowa, Iowa City, Iowa, USA
| | - Devanshee Patel
- Department of Neurology, The University of Iowa Hospitals and Clinics, Iowa City, Iowa, USA
| | - Madhavan L Raghavan
- Roy J Carver Department of Biomedical Engineering, The University of Iowa, Iowa City, Iowa, USA
| | - David Hasan
- Department of Neurosurgery, The University of Iowa Hospitals and Clinics, Iowa City, Iowa, USA
| | - Edgar A Samaniego
- Department of Neurology, The University of Iowa Hospitals and Clinics, Iowa City, Iowa, USA .,Department of Neurosurgery, The University of Iowa Hospitals and Clinics, Iowa City, Iowa, USA.,Department of Radiology, The University of Iowa Hospitals and Clinics, Iowa City, Iowa, USA
| |
Collapse
|
30
|
Hoxha M, Tedesco CC, Quaglin S, Malaj V, Pustina L, Capra V, Evans JF, Sala A, Rovati GE. Montelukast Use Decreases Cardiovascular Events in Asthmatics. Front Pharmacol 2021; 11:611561. [PMID: 33519477 PMCID: PMC7838535 DOI: 10.3389/fphar.2020.611561] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Accepted: 11/12/2020] [Indexed: 11/13/2022] Open
Abstract
Cysteinyl leukotrienes are proinflammatory mediators with a clinically established role in asthma and a human genetic and preclinical role in cardiovascular pathology. Given that cardiovascular disease has a critical inflammatory component, the aim of this work was to conduct an observational study to verify whether the use of a cysteinyl leukotriene antagonist, namely, montelukast, may protect asthmatic patients from a major cardiovascular event and, therefore, represent an innovative adjunct therapy to target an inflammatory component in cardiovascular disease. We performed an observational retrospective 3-year study on eight hundred adult asthmatic patients 18 years or older in Albania, equally distributed into two cohorts, exposed or nonexposed to montelukast usage, matched by age and gender according to information reported in the data collection. Patients with a previous history of myocardial infarction or ischemic stroke were excluded. In summary, 37 (4.6%) of the asthmatic patients, 32 nonexposed, and five exposed to montelukast suffered a major cardiovascular event during the 3-year observation period. All the cardiovascular events, in either group, occurred among patients with an increased cardiovascular risk. Our analyses demonstrate that, independent from gender, exposure to montelukast remained a significant protective factor for incident ischemic events (78% or 76% risk reduction depending on type of analysis). The event-free Kaplan–Meier survival curves confirmed the lower cardiovascular event incidence in patients exposed to montelukast. Our data suggest that there is a potential preventative role of montelukast for incident cardiac ischemic events in the older asthmatic population, indicating a comorbidity benefit of montelukast usage in asthmatics by targeting cysteinyl leukotriene-driven cardiac disease inflammation.
Collapse
Affiliation(s)
- Malvina Hoxha
- Department of Pharmaceutical Sciences, University of Milan, Milano, Italy.,Department for Chemical-Toxicological and Pharmacologicsal Evaluation of Drugs, Catholic University Our Lady of Good Counsel, Tirana, Albania
| | | | - Silvana Quaglin
- Department of Industrial Engineering and Information, University of Pavia, Pavia, Italy
| | - Visar Malaj
- Department of Economics, Faculty of Economics, University of Tirana, Tirana, Albania
| | | | - Valerie Capra
- Department of Pharmaceutical Sciences, University of Milan, Milano, Italy
| | - Jilly F Evans
- University of Pennsylvania School of Medicine, Philadelphia, PA, United States
| | - Angelo Sala
- Department of Pharmaceutical Sciences, University of Milan, Milano, Italy.,IBIM, Consiglio Nazionale Delle Ricerche, Palermo, Italy
| | - G Enrico Rovati
- Department of Pharmaceutical Sciences, University of Milan, Milano, Italy
| |
Collapse
|
31
|
Hoxha M, Malaj V, Vara-Messler M, Doce CR, Cavanillas AB. A case-control study: Evaluating the role of leukotriene receptor antagonists in preventing the cardiovascular and cerebrovascular disease. Semergen 2020; 47:4-11. [PMID: 33277178 DOI: 10.1016/j.semerg.2020.09.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Revised: 08/23/2020] [Accepted: 09/29/2020] [Indexed: 11/30/2022]
Abstract
INTRODUCTION Leukotriene receptor antagonists (LTRAs) are used as a therapeutic alternative in asthmatic patients. Different animal studies indicate that LTRAs can decrease intimal hyperplasia after vascular injury, and have a protective role in cerebral ischemia. OBJECTIVE The aim of this study was to assess the role of leukotriene receptor antagonists in preventing the cardiovascular and ischemic stroke in humans. MATERIAL AND METHOD A matched case-control study with a follow up period of three years has been conducted, investigating the effect of the LTRAs in the myocardial infarct (MI) risk, and in the ischemic stroke (IS) risk in asthmatic patients from San Cecilio University Hospital of Granada, and from two Primary Health Care Centers of Granada. RESULTS 59 cases with MI and 108 cases with IS were included in the study, each of them with an equal number of controls matched by age and sex in each of the two Health Care Centers. Unlike for MI risk, the treatment with LTRAs was associated with a slight trend in reducing the risk of stroke, in both of the primary care controls (Odds ratios: 0.74 (0.37-1.47); 0.82 (0.4-1.67), for the first, and the second Health Centers Controls, respectively), but without reaching a statistical significance. CONCLUSIONS The results did not confirm a protective effect of LTRAs on cardiovascular risk as suggested by different animal studies.
Collapse
Affiliation(s)
- M Hoxha
- Catholic University Our Lady of Good Counsel, Department of Chemical, Toxicological and Pharmacological Evaluation of Drugs, Rruga Dritan Hoxha, Tirana, Albania; Università degli studi di Milano, Department of Pharmacological and Biomolecular Sciences, Via Balzaretti, 9-20133 Milan, Italy.
| | - V Malaj
- University of Tirana, Faculty of Economics, Department of Economics, Rruga Arben Broci, Tirana, Albania
| | - M Vara-Messler
- University of Turin, Department of Oncology, Via Verdi, 8-10124 Turin, Italy
| | - C R Doce
- IBS Granada, University of Granada, Ciber of Epidemiology and Public, Spain
| | - A B Cavanillas
- IBS Granada, University of Granada, Ciber of Epidemiology and Public, Spain
| |
Collapse
|
32
|
Rhee TM, Choi EK, Han KD, Lee SR, Oh S. Impact of the Combinations of Allergic Diseases on Myocardial Infarction and Mortality. THE JOURNAL OF ALLERGY AND CLINICAL IMMUNOLOGY-IN PRACTICE 2020; 9:872-880.e4. [PMID: 32961311 DOI: 10.1016/j.jaip.2020.09.008] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 03/19/2020] [Revised: 08/29/2020] [Accepted: 09/05/2020] [Indexed: 01/31/2023]
Abstract
BACKGROUND Allergic rhinitis (AR), atopic dermatitis (AD), and asthma, each by itself, are known to be associated with a higher risk of cardiovascular disease. Each of these diseases often coexists with one another, but the effect of combined allergic diseases on the long-term risk of myocardial infarction (MI) and mortality remains unknown. OBJECTIVE To evaluate the effects of various combinations of the allergic triad on the risk of MI and mortality. METHODS Adult allergic disease patients without prior MI were enrolled from the nationwide health check-up data provided by the Korean National Health Insurance Service in 2009. The primary and secondary end points were all-cause death and MI. RESULTS A total of 9,548,939 individuals older than 20 years were selected for analysis. The prevalence of allergic diseases was 13.9% for AR, 0.4% for AD, and 2.7% for asthma. During a median 8.2 years of follow-up, 105,659 MIs and 298,769 deaths occurred. All allergic diseases were associated with an increased risk of MI (AR, adjusted hazard ratio [HRadjust], 1.11, 95% CI, 1.10-1.13; AD, HRadjust, 1.14, 95% CI, 1.06-1.24; asthma, HRadjust, 1.37, 95% CI, 1.33-1.40), whereas mortality risk was increased only for patients with AD (HRadjust, 1.15; 95% CI, 1.10-1.20) or asthma (HRadjust, 1.41; 95% CI, 1.39-1.43). Among the combinations of allergic diseases, patients with both AD and asthma had the highest risk of mortality (HRadjust, 1.71; 95% CI, 1.46-2.00) and MI (HRadjust, 1.57; 95% CI, 1.15-2.16). The results were significant after adjusting for demographic characteristics, comorbidities, socioeconomic status, and lifestyle factors. CONCLUSIONS Patients with both AD and asthma have the highest risk of MI and mortality among all allergic disease combinations. Physicians should evaluate combinations of allergic conditions in allergic disease patients and promptly assess and manage their future risk of MI and mortality.
Collapse
Affiliation(s)
- Tae-Min Rhee
- Division of Cardiology, Department of Internal Medicine, Seoul National University Hospital, Seoul, Korea
| | - Eue-Keun Choi
- Department of Statistics and Actuarial Science, Soongsil University, Seoul, Korea.
| | - Kyung-Do Han
- Department of Statistics and Actuarial Science, Soongsil University, Seoul, Korea
| | - So-Ryoung Lee
- Division of Cardiology, Department of Internal Medicine, Seoul National University Hospital, Seoul, Korea
| | - Seil Oh
- Division of Cardiology, Department of Internal Medicine, Seoul National University Hospital, Seoul, Korea
| |
Collapse
|
33
|
Ajala ON, Everett BM. Targeting Inflammation to Reduce Residual Cardiovascular Risk. Curr Atheroscler Rep 2020; 22:66. [DOI: 10.1007/s11883-020-00883-3] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
|
34
|
Tang L, Wang Z, Mu Q, Yu Z, Jacobson O, Li L, Yang W, Huang C, Kang F, Fan W, Ma Y, Wang M, Zhou Z, Chen X. Targeting Neutrophils for Enhanced Cancer Theranostics. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2020; 32:e2002739. [PMID: 32656801 DOI: 10.1002/adma.202002739] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Revised: 06/18/2020] [Indexed: 05/18/2023]
Abstract
Improving tumor accumulation and delivery efficiency is an important goal of nanomedicine. Neutrophils play a vital role in both chemically mediating inflammatory response through myeloperoxidase (MPO) and biologically promoting metastasis during inflammation triggered by the primary tumor or environmental stimuli. Herein, a novel theranostic nanomedicine that targets both the chemical and biological functions of neutrophils in tumor is designed, facilitating the enhanced retention and sustained release of drug cargos for improved cancer theranostics. 5-hydroxytryptamine (5-HT) is equipped onto nanoparticles (NPs) loaded with photosensitizers and Zileuton (a leukotriene inhibitor) to obtain MPO and neutrophil targeting NPs, denoted as HZ-5 NPs. The MPO targeting property of 5-HT modified NPs is confirmed by noninvasive positron emission tomography imaging studies. Furthermore, photodynamic therapy is used to initiate the inflammatory response which further mediated the accumulation and retention of neutrophil targeting NPs in a breast cancer model. This design renders a greatly improved theranostic nanomedicine for efficient tumor suppression, and more importantly, inhibition of neutrophil-mediated lung metastasis via the sustained release of Zileuton. This work presents a novel strategy of targeting neutrophils for improved tumor theranostics, which may open up new avenues in designing nanomedicine through exploiting the tumor microenvironment.
Collapse
Affiliation(s)
- Longguang Tang
- The People's Hospital of Gaozhou, Maoming, 525200, China
- Laboratory of Molecular Imaging and Nanomedicine, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, MD, 20892, USA
- Department of Chemistry, Stanford University, Stanford, CA, 94305, USA
| | - Zhantong Wang
- Laboratory of Molecular Imaging and Nanomedicine, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Qingchun Mu
- The People's Hospital of Gaozhou, Maoming, 525200, China
| | - Zhiqiang Yu
- School of Pharmaceutical Sciences, Guangdong Provincial Key Laboratory of New Drug Screening, Southern Medical University, Guangzhou, 510515, China
| | - Orit Jacobson
- Laboratory of Molecular Imaging and Nanomedicine, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Ling Li
- Laboratory of Molecular Imaging and Nanomedicine, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Weijing Yang
- Laboratory of Molecular Imaging and Nanomedicine, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Chunming Huang
- The People's Hospital of Gaozhou, Maoming, 525200, China
| | - Fei Kang
- Laboratory of Molecular Imaging and Nanomedicine, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Wenpei Fan
- Laboratory of Molecular Imaging and Nanomedicine, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Ying Ma
- Laboratory of Molecular Imaging and Nanomedicine, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Maosheng Wang
- The People's Hospital of Gaozhou, Maoming, 525200, China
| | - Zijian Zhou
- Laboratory of Molecular Imaging and Nanomedicine, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Xiaoyuan Chen
- Laboratory of Molecular Imaging and Nanomedicine, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, MD, 20892, USA
| |
Collapse
|
35
|
Targeted anti-inflammatory therapy is a new insight for reducing cardiovascular events: A review from physiology to the clinic. Life Sci 2020; 253:117720. [PMID: 32360620 DOI: 10.1016/j.lfs.2020.117720] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Revised: 04/13/2020] [Accepted: 04/14/2020] [Indexed: 12/30/2022]
Abstract
Despite considerable progressions, cardiovascular disease (CVD) is still one of the major causes of mortality around the world, indicates an important and unmet clinical need. Recently, extensive studies have been performed on the role of inflammatory factors as either a major or surrogate factor in the pathophysiology of CVD. Epidemiological observations suggest the theory of the role of inflammatory mediators in the development of cardiovascular events. This may support the idea that targeted anti-inflammatory therapies, on the background of traditional validated medical therapies, can play a significant role in prevention and even reduction of cardiovascular disorders. Many randomized controlled trials have shown that drugs commonly useful for primary and secondary prevention of CVD have an anti-inflammatory mechanism. Further, many anti-inflammatory drugs are being examined because of their potential to reduce the risk of cardiovascular problems. In this study, we review the process of inflammation in the development of cardiovascular events, both in vivo and clinical evidence in immunotherapy for CVD.
Collapse
|
36
|
Leuti A, Fazio D, Fava M, Piccoli A, Oddi S, Maccarrone M. Bioactive lipids, inflammation and chronic diseases. Adv Drug Deliv Rev 2020; 159:133-169. [PMID: 32628989 DOI: 10.1016/j.addr.2020.06.028] [Citation(s) in RCA: 212] [Impact Index Per Article: 42.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Revised: 06/09/2020] [Accepted: 06/25/2020] [Indexed: 02/08/2023]
Abstract
Endogenous bioactive lipids are part of a complex network that modulates a plethora of cellular and molecular processes involved in health and disease, of which inflammation represents one of the most prominent examples. Inflammation serves as a well-conserved defence mechanism, triggered in the event of chemical, mechanical or microbial damage, that is meant to eradicate the source of damage and restore tissue function. However, excessive inflammatory signals, or impairment of pro-resolving/anti-inflammatory pathways leads to chronic inflammation, which is a hallmark of chronic pathologies. All main classes of endogenous bioactive lipids - namely eicosanoids, specialized pro-resolving lipid mediators, lysoglycerophopsholipids and endocannabinoids - have been consistently involved in the chronic inflammation that characterises pathologies such as cancer, diabetes, atherosclerosis, asthma, as well as autoimmune and neurodegenerative disorders and inflammatory bowel diseases. This review gathers the current knowledge concerning the involvement of endogenous bioactive lipids in the pathogenic processes of chronic inflammatory pathologies.
Collapse
|
37
|
Almeida SO, Ram RJ, Kinninger A, Budoff MJ. Effect of 5-lipoxygenase inhibitor, VIA-2291 (Atreleuton), on epicardial fat volume in patients with recent acute coronary syndrome. J Cardiovasc Comput Tomogr 2020; 14:343-348. [DOI: 10.1016/j.jcct.2019.12.033] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/24/2019] [Revised: 12/21/2019] [Accepted: 12/24/2019] [Indexed: 12/01/2022]
|
38
|
Löfgren L, Forsberg GB, Davidsson P, Eketjäll S, Whatling C. Development of a highly sensitive liquid chromatography-mass spectrometry method to quantify plasma leukotriene E 4 and demonstrate pharmacological suppression of endogenous 5-LO pathway activity in man. Prostaglandins Other Lipid Mediat 2020; 150:106463. [PMID: 32450304 DOI: 10.1016/j.prostaglandins.2020.106463] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Revised: 04/20/2020] [Accepted: 05/19/2020] [Indexed: 11/15/2022]
Abstract
Low basal endogenous concentrations (<20 pg/mL) of the 5-lipoxygenase (5-LO) pathway biomarker leukotriene E4 (LTE4) in human plasma present a significant analytical challenge. Analytical methods including liquid chromatography-mass spectrometry and enzyme linked immunosorbent assays have been used to quantify plasma LTE4 in the past but have not provided consistent data in the lower pg/mL-range. With our new method, a detection limit (<1 pg/mL plasma) significantly below basal levels of LTE4 was achieved by combining large volume sample purification and enrichment by anion-exchange mixed mode solid phase extraction (SPE) with large volume injection followed by chromatographic separation by ultra performance liquid chromatography (UPLC) and quantification by highly sensitive negative-ion electrospray tandem mass spectrometry (MS/MS). The method was reproducible, accurate and linear between 1 and 120 pg/mL plasma LTE4. The method was used to perform an analysis of plasma samples collected from healthy volunteers in a Phase 1 study with the FLAP (5-lipoxygenase activating protein) inhibitor AZD5718. Basal endogenous LTE4 levels of 5.1 ± 2.7 pg/mL were observed in healthy volunteers (n = 34). In subjects that had been administered a single oral dose of AZD5718, significant suppression (>80%) of plasma LTE4 level was observed, providing pharmacological evidence that endogenous 5-LO pathway activity could be assessed.
Collapse
Affiliation(s)
- Lars Löfgren
- Translational Science & Experimental Medicine, Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden.
| | - Gun-Britt Forsberg
- Translational Science & Experimental Medicine, Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Pia Davidsson
- Translational Science & Experimental Medicine, Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Susanna Eketjäll
- Translational Science & Experimental Medicine, Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Carl Whatling
- Translational Science & Experimental Medicine, Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| |
Collapse
|
39
|
He R, Chen Y, Cai Q. The role of the LTB4-BLT1 axis in health and disease. Pharmacol Res 2020; 158:104857. [PMID: 32439596 DOI: 10.1016/j.phrs.2020.104857] [Citation(s) in RCA: 68] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2020] [Revised: 04/03/2020] [Accepted: 04/20/2020] [Indexed: 12/16/2022]
Abstract
Leukotriene B4 (LTB4) is a major type of lipid mediator that is rapidly generated from arachidonic acid through sequential action of 5-lipoxygenase (5-LO), 5-lipoxygenase-activating protein (FLAP) and LTA4 hydrolase (LTA4H) in response to various stimuli. LTB4 is well known to be a chemoattractant for leukocytes, particularly neutrophils, via interaction with its high-affinity receptor BLT1. Extensive attention has been paid to the role of the LTB4-BLT1 axis in acute and chronic inflammatory diseases, such as infectious diseases, allergy, autoimmune diseases, and metabolic disease via mediating recruitment and/or activation of different types of inflammatory cells depending on different stages or the nature of inflammatory response. Recent studies also demonstrated that LTB4 acts on non-immune cells via BLT1 to initiate and/or amplify pathological inflammation in various tissues. In addition, emerging evidence reveals a complex role of the LTB4-BLT1 axis in cancer, either tumor-inhibitory or tumor-promoting, depending on the different target cells. In this review, we summarize both established understanding and the most recent progress in our knowledge about the LTB4-BLT1 axis in host defense, inflammatory diseases and cancer.
Collapse
Affiliation(s)
- Rui He
- Department of Immunology, School of Basic Medical Sciences, Fudan University, Shanghai, People's Republic of China.
| | - Yu Chen
- Department of Immunology, School of Basic Medical Sciences, Fudan University, Shanghai, People's Republic of China
| | - Qian Cai
- Department of Immunology, School of Basic Medical Sciences, Fudan University, Shanghai, People's Republic of China
| |
Collapse
|
40
|
Comprehensive plaque assessment with serial coronary CT angiography: translation to bedside. Int J Cardiovasc Imaging 2020; 36:2335-2346. [DOI: 10.1007/s10554-020-01849-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Accepted: 04/09/2020] [Indexed: 12/24/2022]
|
41
|
Cilenšek I, Šeruga M, Makuc J, Završnik M, Petrovič D. The ALOXA5AP gene (rs38022789) is associated with diabetic nephropathy in Slovenian patients with type 2 diabetes mellitus. Gene 2020; 741:144551. [PMID: 32165305 DOI: 10.1016/j.gene.2020.144551] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2019] [Revised: 10/08/2019] [Accepted: 03/08/2020] [Indexed: 01/11/2023]
Abstract
BACKGROUND Although the pathogenesis of diabetic nephropathy (DN) is multifactorial, and the precise mechanisms are unclear, there is a growing body of evidence suggesting that inflammatory processes and immune cells might be involved in the development and progression of DN. Leukotrienes (LTs) are a family of lipid mediators, which act as pro-inflammatory mediators. The study was designed to investigate the association between the polymorphism of the ALOX5 gene (rs12762303) and the ALOX5AP gene (rs3802278), and DN in patients with T2DM. METHODOLOGY 651 subjects with diabetes mellitus type 2 (T2DM) were classified into two groups according to the presence of DN, and tested for ALOX5 and ALOX5AP gene polymorphisms using the KASPar genotyping chemistry with validated assay. Biochemical analyses were performed using standard biochemical methods. RESULTS Logistic regression analysis demonstrated that the carriers of the CC genotype had a 3.14 higher risk for DN compared to TT genotype. Serum cystatin C was found to be statistically significantly higher in cases with DN in comparison with subjects without DN (p < 0.001). CONCLUSION An association between the rs3803278 of the ALOX5AP gene and DN was found in Slovenian patients with T2DM. The rs3803278 CC allele appears to confer increased risk of DN possibly by increasing the production of LTs-potent drivers of inflammation.
Collapse
Affiliation(s)
- Ines Cilenšek
- Institute of Histology and Embryology, Faculty of Medicine, University of Ljubljana, Slovenia.
| | - Maja Šeruga
- Department of Internal Medicine, General Hospital Murska Sobota, Murska Sobota, Slovenia
| | - Jana Makuc
- Department of Internal Medicine, General Hospital Slovenj Gradec, Slovenj Gradec, Slovenia
| | - Matej Završnik
- Department for Diabetes and Metabolic Diseases, Clinic for Internal Medicine, University Medical Centre Maribor, Maribor, Slovenia
| | - Danijel Petrovič
- Institute of Histology and Embryology, Faculty of Medicine, University of Ljubljana, Slovenia
| |
Collapse
|
42
|
Chan SJ, Ng MPE, Zhao H, Ng GJL, De Foo C, Wong PTH, Seet RCS. Early and Sustained Increases in Leukotriene B 4 Levels Are Associated with Poor Clinical Outcome in Ischemic Stroke Patients. Neurotherapeutics 2020; 17:282-293. [PMID: 31520306 PMCID: PMC7007445 DOI: 10.1007/s13311-019-00787-4] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Leukotriene B4 (LTB4) has been implicated in ischemic stroke pathology. We examined the prognostic significance of LTB4 levels in patients with acute middle cerebral artery (MCA) infarction and their mechanisms in rat stroke models. In ischemic stroke patients with middle cerebral artery infarction, plasma LTB4 levels were found to increase rapidly, roughly doubling within 24 h when compared to initial post-stroke levels. Further analyses indicate that poor functional recovery is associated with early and more sustained increase in LTB4 rather than the peak levels. Results from studies using a rat embolic stroke model showed increased 5-lipoxygenase (5-LOX) expression in the ipsilateral infarcted cortex compared with sham control or respective contralateral regions at 24 h post-stroke with a concomitant increase in LTB4 levels. In addition, neutrophil influx was also observed in the infarcted cortex. Double immunostaining indicated that neutrophils express 5-LOX and leukotriene A4 hydrolase (LTA4H), highlighting the pivotal contributions of neutrophils as a source of LTB4. Importantly, rise in plasma LTB4 levels corresponded with an increase in LTB4 amount in the infarcted cortex, thereby supporting the use of plasma as a surrogate for brain LTB4 levels. Pre-stroke LTB4 loading increased brain infarct volume in tMCAO rats. Conversely, administration of the 5-LOX-activating protein (FLAP) inhibitor BAY-X1005 or B-leukotriene receptor (BLTR) antagonist LY255283 decreased the infarct volume by a similar extent. To conclude, targeted interruption of the LTB4 pathway might be a viable treatment strategy for acute ischemic stroke.
Collapse
Affiliation(s)
- Su Jing Chan
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University Health System, National University of Singapore, MD3, 16 Medical Drive, Singapore, 117600, Singapore
| | - Mary P E Ng
- Department of Medicine, Yong Loo Lin School of Medicine, National University Health System, National University of Singapore, NUHS Tower Block, 1E Kent Ridge Road, Singapore, 119228, Singapore
| | - Hui Zhao
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University Health System, National University of Singapore, MD3, 16 Medical Drive, Singapore, 117600, Singapore
| | - Geelyn J L Ng
- Department of Medicine, Yong Loo Lin School of Medicine, National University Health System, National University of Singapore, NUHS Tower Block, 1E Kent Ridge Road, Singapore, 119228, Singapore
| | - Chuan De Foo
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University Health System, National University of Singapore, MD3, 16 Medical Drive, Singapore, 117600, Singapore
| | - Peter T-H Wong
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University Health System, National University of Singapore, MD3, 16 Medical Drive, Singapore, 117600, Singapore.
| | - Raymond C S Seet
- Department of Medicine, Yong Loo Lin School of Medicine, National University Health System, National University of Singapore, NUHS Tower Block, 1E Kent Ridge Road, Singapore, 119228, Singapore.
| |
Collapse
|
43
|
Camacho-Mejorado R, Gómez R, Torres-Sánchez LE, Alhelí Hernández-Tobías E, Noris G, Santana C, Magaña JJ, Orozco L, de la Peña-Díaz A, de la Luz Arenas-Sordo M, Meraz-Ríos MA, Majluf-Cruz A. ALOX5, LPA, MMP9 and TPO gene polymorphisms increase atherothrombosis susceptibility in middle-aged Mexicans. ROYAL SOCIETY OPEN SCIENCE 2020; 7:190775. [PMID: 32218930 PMCID: PMC7029922 DOI: 10.1098/rsos.190775] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/10/2019] [Accepted: 11/28/2019] [Indexed: 06/10/2023]
Abstract
Atherothrombosis is the cornerstone of cardiovascular diseases and the primary cause of death worldwide. Genetic contribution to disturbances in lipid metabolism, coagulation, inflammation and oxidative stress increase the susceptibility to its development and progression. Given its multifactorial nature, the multiloci studies have been proposed as potential predictors of susceptibility. A cross-sectional study was conducted to explore the contribution of nine genes involved in oxidative stress, inflammatory and thrombotic processes in 204 subjects with atherothrombosis matched by age and gender with a healthy group (n = 204). To evaluate the possibility of spurious associations owing to the Mexican population genetic heterogeneity as well as its ancestral origins, 300 unrelated mestizo individuals and 329 Native Americans were also included. ALOX5, LPA, MMP9 and TPO gene polymorphisms, as well as their multiallelic combinations, were twice to four times more frequent in those individuals with clinical manifestations of atherothrombosis than in the healthy group. Once adjusting for population stratification was done, these differences remained. Our results add further evidence on the contribution of ALOX5, LPA, MMP9 and TPO polymorphisms to atherothrombosis development in the middle-aged group, emphasizing the multiethnic studies in search of gene risk polymorphisms.
Collapse
Affiliation(s)
| | - Rocío Gómez
- Departamento de Toxicología, Cinvestav-IPN, Mexico City 07360, Mexico
| | - Luisa E. Torres-Sánchez
- Centro de Investigación en Salud Poblacional, Instituto Nacional de Salud Pública, Cuernavaca, Morelos, Mexico
| | | | - Gino Noris
- Laboratorio Biología Molecular Diagnóstica, Querétaro, Qro, Mexico
| | - Carla Santana
- Laboratorio Biología Molecular Diagnóstica, Querétaro, Qro, Mexico
| | | | - Lorena Orozco
- Laboratorio de Inmunogenómica y Enfermedades Metabólicas, INMEGEN, Mexico City, Mexico
| | - Aurora de la Peña-Díaz
- Facultad de Medicina, Departamento de Farmacología, Universidad Nacional Autónoma de México, Mexico
- Departamento de Biología Molecular, Instituto Nacional de Cardiología, Mexico City, Mexico
| | | | | | - Abraham Majluf-Cruz
- Unidad de Investigación Médica en Trombosis, Hemostasia y Aterogénesis, IMSS, Mexico City, Mexico
| |
Collapse
|
44
|
Abstract
Background:
Exposure to air pollution increases cardiovascular morbidity and mortality. Preventing chronic cardiovascular diseases caused by air pollution relies on detecting the early effects of pollutants on the risk of cardiovascular disease development, which is limited by the lack of sensitive biomarkers. We have previously identified promising biomarkers in experimental animals but comparable evidence in humans is lacking.
Methods:
Air pollution is substantially worse in Beijing than in Los Angeles. We collected urine and blood samples from 26 nonsmoking, healthy adult residents of Los Angeles (mean age, 23.8 years; 14 women) before, during, and after spending 10 weeks in Beijing during the summers of 2014 and 2015. We assessed a panel of circulating biomarkers indicative of lipid peroxidation and inflammation. Personal exposure to polycyclic aromatic hydrocarbons (PAHs), a group of combustion-originated air pollutants, was assessed by urinary PAH metabolite levels.
Results:
Urinary concentrations of 4 PAH metabolites were 176% (95% CI, 103% to 276%) to 800% (95% CI, 509% to 1780%) greater in Beijing than in Los Angeles. Concentrations of 6 lipid peroxidation biomarkers were also increased in Beijing, among which 5-, 12-, and 15-hydroxyeicosatetraenoic acid and 9- and 13-hydroxyoctadecadienoic acid levels reached statistical significance (false discovery rate <5%), but not 8-isoprostane (20.8%; 95% CI, −5.0% to 53.6%). The antioxidative activities of paraoxonase (−9.8%; 95% CI, −14.0% to −5.3%) and arylesterase (−14.5%; 95% CI, −22.3% to −5.8%) were lower and proinflammatory C-reactive protein (101%; 95% CI, 3.3% to 291%) and fibrinogen (48.3%; 95% CI, 4.9% to 110%) concentrations were higher in Beijing. Changes in all these biomarkers were reversed, at least partially, after study participants returned to Los Angeles. Changes in most outcomes were associated with urinary PAH metabolites (
P
<0.05).
Conclusions:
Traveling from a less-polluted to a more-polluted city induces systemic pro-oxidative and proinflammatory effects. Changes in the levels of 5-, 12-, and 15-hydroxyeicosatetraenoic acid and 9- and 13-hydroxyoctadecadienoic acid as well as paraoxonase and arylesterase activities in the blood, in association with exposures to PAH metabolites, might have important implications in preventive medicine as indicators of increased cardiovascular risk caused by air pollution exposure.
Collapse
|
45
|
Ericsson H, Nelander K, Heijer M, Kjaer M, Lindstedt EL, Albayaty M, Forte P, Lagerström-Fermér M, Skrtic S. Phase 1 Pharmacokinetic Study of AZD5718 in Healthy Volunteers: Effects of Coadministration With Rosuvastatin, Formulation and Food on Oral Bioavailability. Clin Pharmacol Drug Dev 2019; 9:411-421. [PMID: 31793171 PMCID: PMC7187334 DOI: 10.1002/cpdd.756] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2019] [Accepted: 11/05/2019] [Indexed: 12/21/2022]
Abstract
AZD5718 is a first‐in‐class small‐molecule anti‐inflammatory drug with the potential to reduce the residual risk of cardiovascular events after myocardial infarction in patients receiving lipid‐lowering statin therapy. Leukotrienes are potent proinflammatory and vasoactive mediators synthesized in leukocytes via 5‐lipoxygenase and 5‐lipoxygenase‐activating protein (FLAP). AZD5718 is a FLAP inhibitor that dose‐dependently reduced leukotriene biosynthesis in a first‐in‐human study. We enrolled 12 healthy men in a randomized, open‐label, crossover, single‐dose phase 1 pharmacokinetic study of AZD5718 to investigate a potential drug‐drug interaction with rosuvastatin, and the effects of formulation and food intake (ClinicalTrials.gov identifier: NCT02963116). Rosuvastatin (10 mg) were absorbed more rapidly when coadministered with AZD5718 (200 mg), probably owing to weak inhibition of hepatic statin uptake, but relative bioavailability was unaffected (geometric least‐squares mean ratio [GMR], 100%; 90% confidence interval [CI], 86%‐116%). AZD5718 pharmacokinetics were unaffected by coadministration of rosuvastatin. AZD5718 (200 mg) was absorbed less rapidly when formulated as tablets than oral suspension, with reduced relative bioavailability (GMR, 72%; 90%CI, 64%‐80%). AZD5718 absorption was slower when 200‐mg tablets were taken after a high‐fat breakfast than after fasting, but relative bioavailability was unaffected (GMR, 96%; 90%CI, 87%‐106%). In post hoc pharmacodynamic simulations, plasma leukotriene B4 levels were inhibited by >90% throughout the day following once‐daily AZD5718, regardless of formulation or administration with food. AZD5718 was well tolerated, with no severe or serious adverse events. These data supported the design of a phase 2a efficacy study of AZD5718 in patients with coronary artery disease.
Collapse
Affiliation(s)
- Hans Ericsson
- Clinical Pharmacology, ADME and AI, Clinical Pharmacology & Safety Sciences, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Karin Nelander
- Clinical Pharmacology, ADME and AI, Clinical Pharmacology & Safety Sciences, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Maria Heijer
- Clinical Pharmacology Biologics and Bioanalysis, Clinical Pharmacology & Safety Sciences, R&D, AstraZeneca, Gothenburg, Sweden
| | - Magnus Kjaer
- Early Biometrics and Statistical Innovation, Data Science & AI, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Eva-Lotte Lindstedt
- Research and Early Development, Cardiovascular, Renal and Metabolic, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | | | - Pablo Forte
- Parexel, Early Phase Clinical Unit, Harrow, UK
| | - Maria Lagerström-Fermér
- Research and Early Development, Cardiovascular, Renal and Metabolic, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Stanko Skrtic
- Research and Early Development, Cardiovascular, Renal and Metabolic, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden.,Department of Internal Medicine and Clinical Nutrition, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
46
|
Darwesh AM, Sosnowski DK, Lee TYT, Keshavarz-Bahaghighat H, Seubert JM. Insights into the cardioprotective properties of n-3 PUFAs against ischemic heart disease via modulation of the innate immune system. Chem Biol Interact 2019; 308:20-44. [DOI: 10.1016/j.cbi.2019.04.037] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Revised: 04/17/2019] [Accepted: 04/30/2019] [Indexed: 12/19/2022]
|
47
|
Röhrig T, Kirsch V, Schipp D, Galan J, Richling E. Absorption of Anthocyanin Rutinosides after Consumption of a Blackcurrant ( Ribes nigrum L.) Extract. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2019; 67:6792-6797. [PMID: 31134806 DOI: 10.1021/acs.jafc.9b01567] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
The dominant anthocyanins in blackcurrant are delphinidin-3-O-rutinoside and cyanidin-3-O-rutinoside. Data on their absorption and distribution in the human body are limited. Therefore, we performed a human pilot study on five healthy male volunteers consuming a blackcurrant ( Ribes nigrum L.) extract. The rutinosides and their degradation products gallic acid and protocatechuic acid were determined in plasma and urine. The rutinosides' concentrations peaked in both plasma and urine samples within 2 h of extract ingestion. The recoveries of delphinidin-3-O-rutinoside and cyanidin-3-O-rutinoside from urine samples were 0.040 ± 0.011% and 0.048 ± 0.016%, respectively, over a 48 h period. Protocatechuic acid concentration increased significantly after ingestion of the blackcurrant extract. Our results show that after ingestion of a blackcurrant extract containing delphinidin-3-O-rutinoside and cyanidin-3-O-rutinoside, significant quantities of biologically active compounds circulated in the plasma and were excreted via urine. Furthermore, these results contribute to the understanding of anthocyanin metabolism in humans.
Collapse
Affiliation(s)
- Teresa Röhrig
- Division of Food Chemistry and Toxicology, Department of Chemistry , Technische Universitaet Kaiserslautern , Erwin-Schroedinger-Strasse 52 , D-67663 Kaiserslautern , Germany
| | - Verena Kirsch
- Division of Food Chemistry and Toxicology, Department of Chemistry , Technische Universitaet Kaiserslautern , Erwin-Schroedinger-Strasse 52 , D-67663 Kaiserslautern , Germany
| | - Dorothea Schipp
- ds-statistik.de , Pirnaer Strasse 1 , 01824 Rosenthal-Bielatal , Germany
| | - Jens Galan
- Specialist in Inner & General Medicine , Hochgewanne 19 , 67269 Gruenstadt , Germany
| | - Elke Richling
- Division of Food Chemistry and Toxicology, Department of Chemistry , Technische Universitaet Kaiserslautern , Erwin-Schroedinger-Strasse 52 , D-67663 Kaiserslautern , Germany
| |
Collapse
|
48
|
Highly sensitive and specific LC–MS/MS method to determine endogenous leukotriene B4 levels in human plasma. Bioanalysis 2019; 11:1055-1066. [DOI: 10.4155/bio-2019-0021] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Aim: To develop a high sensitivity and specific analytical method to measure endogenous levels of leukotriene B4 (LTB4) in human plasma. Methodology: LC–MS/MS and ELISA. Results: An LC–MS/MS method was developed with a sensitivity of 1.0 pg/ml, and within and between batch precision of <16% and <13% RSD, respectively. Conclusion: We have developed a sensitive LC–MS/MS method that can detect endogenous LTB4 in human plasma. The LC–MS/MS method displayed correlation with a commercial LTB4 ELISA when analyzing in ex vivo ionophore-stimulated blood samples. For untreated plasma this correlation was lost. Endogenous LTB4 was shown to be unstable in plasma during storage at -20°C and subject to stereoisomer formation. Neither of the assays could quantify endogenous plasma LTB4 in samples stored for long term.
Collapse
|
49
|
Poston RN. Atherosclerosis: integration of its pathogenesis as a self-perpetuating propagating inflammation: a review. Cardiovasc Endocrinol Metab 2019; 8:51-61. [PMID: 31588428 PMCID: PMC6738649 DOI: 10.1097/xce.0000000000000172] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2019] [Accepted: 03/29/2019] [Indexed: 12/22/2022]
Abstract
This review proposes that the development of the atherosclerotic plaque is critically dependent on its inflammatory components forming a self-perpetuating and propagating positive feedback loop. The components involved are: (1) LDL oxidation, (2) activation of the endothelium, (3) recruitment of inflammatory monocytes, (4) macrophage accumulation, which induces LDL oxidation, and (5) macrophage generation of inflammatory mediators, which also activate the endothelium. Through these stages, the positive feedback loop is formed, which generates and promotes expansion of the atherosclerotic process. To illustrate this dynamic of lesion development, the author previously produced a computer simulation, which allowed realistic modelling. This hypothesis on atherogenesis can explain the existence and characteristic focal morphology of the atherosclerotic plaque. Each of the components contributing to the feedback loop is discussed. Many of these components also contain subsidiary positive feedback loops, which could exacerbate the overall process.
Collapse
Affiliation(s)
- Robin N. Poston
- Centre for Microvascular Research, William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| |
Collapse
|
50
|
Jang EJ, Baek SE, Kim EJ, Park SY, Kim CD. HMGB1 enhances AGE-mediated VSMC proliferation via an increase in 5-LO-linked RAGE expression. Vascul Pharmacol 2019; 118-119:106559. [PMID: 30954689 DOI: 10.1016/j.vph.2019.04.001] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2018] [Revised: 01/28/2019] [Accepted: 04/02/2019] [Indexed: 01/11/2023]
Abstract
Receptors for advanced glycation end-product (RAGE) play a pivotal role in the progression of proliferative vascular diseases. However, the precise mechanisms regulating RAGE expression in vascular smooth muscle cells (VSMCs) of the injured vasculatures is unclear. Given the potential importance of 5-lipoxygenase (5-LO) derived mediators in cellular responses mediated by RAGE, this study aimed to evaluate in VSMCs treated with high mobility group box 1 (HMGB1): 1) the RAGE expression; 2) the AGE-induced VSMC proliferation; 3) the role of 5-LO signaling in HMGB1-induced RAGE expression. In cultured human VSMCs stimulated with HMGB1 (100 ng/ml), RAGE mRNA and protein expression were markedly increased along with an increase in AGE-mediated VSMC proliferation. Both of these effects were markedly attenuated in cells pretreated with zileuton (1-10 μM), a 5-LO inhibitor, as well as in cells transfected with 5-LO siRNA, suggesting a potential involvement of 5-LO signaling in HMGB1-mediated RAGE expression in VSMCs. Moreover, 5-LO expression, accompanied by production of leukotrienes was markedly increased in HMGB1-stimulated VSMCs, which was attenuated in cells deficient of TLR2 or RAGE. Taken together, our results suggest that HMGB1-induced increase in 5-LO expression enhances RAGE expression in VSMCs, which stimulates AGE-mediated VSMC proliferation. Thus, the 5-LO-RAGE signaling axis in VSMCs might serve as a potential therapeutic target for vascular remodeling in the injured vasculature.
Collapse
Affiliation(s)
- Eun Jeong Jang
- Department of Pharmacology, School of Medicine, Pusan National University, Yangsan, Gyeongnam 50612, Republic of Korea; Gene & Cell Therapy Research Center for Vessel-associated Diseases, Pusan National University, Yangsan, Gyeongnam 50612, Republic of Korea
| | - Seung Eun Baek
- Department of Pharmacology, School of Medicine, Pusan National University, Yangsan, Gyeongnam 50612, Republic of Korea; Gene & Cell Therapy Research Center for Vessel-associated Diseases, Pusan National University, Yangsan, Gyeongnam 50612, Republic of Korea
| | - Eun Jung Kim
- Department of Pharmacology, School of Medicine, Pusan National University, Yangsan, Gyeongnam 50612, Republic of Korea; Gene & Cell Therapy Research Center for Vessel-associated Diseases, Pusan National University, Yangsan, Gyeongnam 50612, Republic of Korea
| | - So Youn Park
- Department of Pharmacology, School of Medicine, Pusan National University, Yangsan, Gyeongnam 50612, Republic of Korea; Gene & Cell Therapy Research Center for Vessel-associated Diseases, Pusan National University, Yangsan, Gyeongnam 50612, Republic of Korea
| | - Chi Dae Kim
- Department of Pharmacology, School of Medicine, Pusan National University, Yangsan, Gyeongnam 50612, Republic of Korea; Gene & Cell Therapy Research Center for Vessel-associated Diseases, Pusan National University, Yangsan, Gyeongnam 50612, Republic of Korea; Research Institute for Convergence of Biomedical Science and Technology, Pusan National University Yangsan Hospital, Gyeongnam 50612, Republic of Korea.
| |
Collapse
|