51
|
Moore KJ, Koplev S, Fisher EA, Tabas I, Björkegren JLM, Doran AC, Kovacic JC. Macrophage Trafficking, Inflammatory Resolution, and Genomics in Atherosclerosis: JACC Macrophage in CVD Series (Part 2). J Am Coll Cardiol 2019; 72:2181-2197. [PMID: 30360827 DOI: 10.1016/j.jacc.2018.08.2147] [Citation(s) in RCA: 127] [Impact Index Per Article: 25.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/07/2018] [Revised: 07/13/2018] [Accepted: 08/03/2018] [Indexed: 12/31/2022]
Abstract
Atherosclerosis is characterized by the retention of modified lipoproteins in the arterial wall. These modified lipoproteins activate resident macrophages and the recruitment of monocyte-derived cells, which differentiate into mononuclear phagocytes that ingest the deposited lipoproteins to become "foam cells": a hallmark of this disease. In this Part 2 of a 4-part review series covering the macrophage in cardiovascular disease, we critically review the contributions and relevant pathobiology of monocytes, macrophages, and foam cells as relevant to atherosclerosis. We also review evidence that via various pathways, a failure of the resolution of inflammation is an additional key aspect of this disease process. Finally, we consider the likely role played by genomics and biological networks in controlling the macrophage phenotype in atherosclerosis. Collectively, these data provide substantial insights on the atherosclerotic process, while concurrently offering numerous molecular and genomic candidates that appear to hold great promise for selective targeting as clinical therapies.
Collapse
Affiliation(s)
- Kathryn J Moore
- Department of Medicine, Leon H. Charney Division of Cardiology, Marc and Ruti Bell Vascular Biology and Disease Program, New York University School of Medicine, New York, New York
| | - Simon Koplev
- Department of Genetics & Genomic Sciences, Institute of Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Edward A Fisher
- Department of Medicine, Leon H. Charney Division of Cardiology, Marc and Ruti Bell Vascular Biology and Disease Program, New York University School of Medicine, New York, New York
| | - Ira Tabas
- Department of Medicine, Columbia University, New York, New York; Department of Pathology and Cell Biology, Columbia University, New York, New York; Department of Physiology, Columbia University, New York, New York
| | - Johan L M Björkegren
- Department of Genetics & Genomic Sciences, Institute of Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, New York; Integrated Cardio Metabolic Centre, Department of Medicine, Karolinska Institutet, Karolinska Universitetssjukhuset, Huddinge, Sweden
| | - Amanda C Doran
- Department of Medicine, Columbia University, New York, New York
| | - Jason C Kovacic
- The Zena and Michael A. Wiener Cardiovascular Institute, Icahn School of Medicine at Mount Sinai, New York, New York.
| |
Collapse
|
52
|
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: 23] [Impact Index Per Article: 4.6] [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]
|
53
|
Feng C, Yang Y, Yang S, Tu X, Wang Y, Song Y, Hui R, Zhang W. Effect of gene-gene and gene-environment interaction on the risk of first-ever stroke and poststroke death. Mol Genet Genomic Med 2019; 7:e846. [PMID: 31293107 PMCID: PMC6687633 DOI: 10.1002/mgg3.846] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2019] [Revised: 05/13/2019] [Accepted: 06/18/2019] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Multiple genetic and environmental factors contribute to the individual-level heterogeneity in stroke. This study aimed to assess how the genetic interactions confer risk of stroke. METHODS In a Chinese case-control study including 1,405 strokes and 1,263 controls who were followed up (range, 0.1-6.0 years), eight genes, including apolipoprotein(a) (APOA1), methylenetetrahydrofolate reductase (MTHFR), vitamin K epoxide reductase complex subunit 1 (VKORC1), arachidonate 5-lipoxygenase-activating protein (ALOX5AP), NOTCH3, chromosome 9p21.3(Chr.9p21.3), vascular endothelial growth factor (VEGFA), and kinase insert domain-containing receptor (KDR), were analyzed for interactions by the generalized multifactor dimensionality reduction method and validated by the multivariate logistic regression models. The genetic associations with carotid artery intima-media thickness (IMT) were examined. RESULTS The interaction of VKORC1 and Chr.9p21.3 was identified for stroke and its worse prognosis, and subjects having the VKORC1 rs2359612C and Chr.9p21.3 rs10757274G alleles had higher risks for stroke (OR = 1.83, 95% CI = 1.32-2.52) as well as for stroke recurrence (HR = 1.84, 95% CI = 1.24-2.73), cardiovascular events (HR = 1.65, 95% CI = 1.15-2.38), and cardiovascular mortality (HR = 2.16, 95% CI = 1.24-3.79). Supporting, they were associated with higher IMT. Hypertension or physical inactivity increased the risk effect. The interaction of VEGFA rs833061C and KDR rs2305948T was identified for hemorrhagic stroke. CONCLUSIONS Our findings identified two novel genetic interactions of VKORC1 and Chr.9p21.3 and of VEGFA and KDR for risk of stroke and subtypes as well as future stroke prognosis.
Collapse
Affiliation(s)
- Congrui Feng
- Beijing Institute for Brain Disorders, Center for Brain Disorders Research, Capital Medical University, Beijing, China.,State Key Laboratory of Cardiovascular Disease, Hypertension Center, FuWai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences (CAMS) & Peking Union Medical College (PUMC), Beijing, China
| | - Yunyun Yang
- State Key Laboratory of Cardiovascular Disease, Hypertension Center, FuWai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences (CAMS) & Peking Union Medical College (PUMC), Beijing, China
| | - Shujun Yang
- State Key Laboratory of Cardiovascular Disease, Hypertension Center, FuWai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences (CAMS) & Peking Union Medical College (PUMC), Beijing, China
| | - Xin Tu
- College of Life Science and Technology, Human Genome Research Center, Huazhong University of Science and Technology, Hubei, China
| | - Yibo Wang
- State Key Laboratory of Cardiovascular Disease, Hypertension Center, FuWai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences (CAMS) & Peking Union Medical College (PUMC), Beijing, China
| | - Yiqing Song
- Department of Epidemiology, Indiana University Richard M. Fairbanks School of Public Health, Indianapolis, IN
| | - Rutai Hui
- State Key Laboratory of Cardiovascular Disease, Hypertension Center, FuWai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences (CAMS) & Peking Union Medical College (PUMC), Beijing, China
| | - Weili Zhang
- State Key Laboratory of Cardiovascular Disease, Hypertension Center, FuWai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences (CAMS) & Peking Union Medical College (PUMC), Beijing, China
| |
Collapse
|
54
|
Lauretti E, Praticò D. Novel Key Players in the Development of Tau Neuropathology: Focus on the 5-Lipoxygenase. J Alzheimers Dis 2019; 64:S481-S489. [PMID: 29758943 DOI: 10.3233/jad-179931] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Tauopathies belong to a large group of neurodegenerative diseases characterized by progressive accumulation of hyperphosphorylated tau. Tau is a microtubule binding protein which is necessary for their assembly and stability. However, tau affinity for microtubules mainly depends on its phosphorylation status, which is the result of a delicate balance between kinases and phosphatases activity. Any significant changes in this equilibrium can promote tau fibrillation, aggregation, neuronal dysfunction, and ultimately neuronal loss. Despite intensive research, the molecular mechanism(s) leading to tau hyperphosphorylation are still unknown and there is no cure for these diseases. Development of an effective strategy that successfully prevents tau excessive phosphorylation and/or tau aggregation may offer a real therapeutic opportunity for these less investigated neurodegenerative conditions. Beside tau, chronic brain inflammation is a common feature of all tauopathies and 5-lipoxygenase, an inflammatory enzyme, is upregulated in brain regions affected by tau pathology. Recently, in vitro studies and preclinical investigations with animal models of tauopathy have implicated 5-lipoxygenase in the regulation of tau phosphorylation through activation of the cyclin-dependent kinase 5 pathway, supporting the novel hypothesis that this protein is a promising therapeutic target for the treatment of tauopathies. In this article, we will discuss the contribution of the 5-lipoxygenase signaling pathway in the development of tau neuropathology, and the promising potential that drugs targeting this enzyme activation hold as a novel disease-modifying therapeutic approach for tauopathies.
Collapse
Affiliation(s)
- Elisabetta Lauretti
- Alzheimer's Center at Temple, Department of Pharmacology, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA
| | - Domenico Praticò
- Alzheimer's Center at Temple, Department of Pharmacology, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA
| |
Collapse
|
55
|
De Vilder EYG, Cardoen S, Hosen MJ, Le Saux O, De Zaeytijd J, Leroy BP, De Reuck J, Coucke PJ, De Paepe A, Hemelsoet D, Vanakker OM. Pathogenic variants in the ABCC6 gene are associated with an increased risk for ischemic stroke. Brain Pathol 2019; 28:822-831. [PMID: 29722917 DOI: 10.1111/bpa.12620] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2018] [Accepted: 04/29/2018] [Indexed: 12/11/2022] Open
Abstract
Ischemic stroke causes a high mortality and morbidity worldwide. It results from a complex interplay of incompletely known environmental and genetic risk factors. We investigated the ABCC6 gene as a candidate risk factor for ischemic stroke because of the increased ischemic stroke incidence in the autosomal recessive disorder pseudoxanthoma elasticum, caused by biallelic pathogenic ABCC6 variants, the higher cardiovascular risk in heterozygous carriers and the established role of ABCC6 dysfunction in myocardial ischemia. We established segregation of a known pathogenic ABCC6 variant (p.[Arg1314Gln]) in 11/19 family members of an ischemic stroke patient in a large multigenerational family suffering from ischemic stroke and/or cardiovascular disease at a relatively young age. In an independent case-control study in 424 ischemic stroke patients and 250 healthy controls, pathogenic ABCC6 variants were 4.9 times more frequent (P = 0.036; 95% CI 1.11-21.33) in the ischemic stroke patient cohort. To study cellular consequences of ABCC6 deficiency in the brain, immunostaining of brain sections in Abcc6-deficient mice and wild-type controls were performed. An upregulation of Bmp4 and Eng and a downregulation of Alk2 was identified in Abcc6-/- mice, suggesting an increase in apoptosis and angiogenesis. As both of these processes are induced in ischemia, we propose that a pro-ischemic state may explain the higher risk to suffer from ischemic stroke in patients carrying a pathogenic ABCC6 variant, as this may lower the threshold to develop acute ischemic events in these patients. In conclusion, this study identified heterozygous ABCC6 variants as a risk factor for ischemic stroke. Further, dysregulation of Bmp (Bmp4, Alk2) and Tgfβ (Eng) signaling in the brain of Abcc6-/- mice could lead to a pro-ischemic state, lowering the threshold to develop acute ischemic events. These data demonstrate the importance of a molecular analysis of the ABCC6 gene in patients diagnosed with cryptogenic ischemic stroke.
Collapse
Affiliation(s)
- Eva Y G De Vilder
- Center for Medical Genetics, Ghent University Hospital, Ghent, Belgium.,Department of Ophthalmology, Ghent University Hospital, Ghent, Belgium.,Research Foundation - Flanders, Brussels, Belgium
| | - Stefanie Cardoen
- Department of Neurology, Ghent University Hospital, Ghent, Belgium
| | - Mohammad J Hosen
- Department of Genetic Engineering and Biotechnology, Shahjalal University of Science and Technology, Sylhet 3114, Bangladesh
| | - Olivier Le Saux
- Department of Cell and Molecular Biology, The John A. Burns School of Medicine, University of Hawai'i, Honolulu, HI
| | - Julie De Zaeytijd
- Department of Ophthalmology, Ghent University Hospital, Ghent, Belgium
| | - Bart P Leroy
- Center for Medical Genetics, Ghent University Hospital, Ghent, Belgium.,Department of Ophthalmology, Ghent University Hospital, Ghent, Belgium.,Division of Ophthalmology, The Children's Hospital of Philadelphia, Philadelphia, PA
| | - Jacques De Reuck
- Department of Neurology, Ghent University Hospital, Ghent, Belgium
| | - Paul J Coucke
- Center for Medical Genetics, Ghent University Hospital, Ghent, Belgium
| | - Anne De Paepe
- Center for Medical Genetics, Ghent University Hospital, Ghent, Belgium
| | | | | |
Collapse
|
56
|
Li J, Wu Z, Zheng D, Sun Y, Wang S, Yan Y. Bioinformatics analysis of the regulatory lncRNA‑miRNA‑mRNA network and drug prediction in patients with hypertrophic cardiomyopathy. Mol Med Rep 2019; 20:549-558. [PMID: 31180540 PMCID: PMC6579968 DOI: 10.3892/mmr.2019.10289] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2018] [Accepted: 04/25/2019] [Indexed: 01/10/2023] Open
Abstract
Hypertrophic cardiomyopathy (HCM) is a complex inherited cardiovascular disease. The present study investigated the long noncoding (lnc)RNA/microRNA (mi)RNA/mRNA expression pattern of patients with HCM and aimed to identify key molecules involved in the development of this condition. An integrated strategy was conducted to identify differentially expressed miRNAs (DEmiRs), differentially expressed lncRNAs (DElncs) and differentially expressed genes (DEGs) based on the GSE36961 (mRNA), GSE36946 (miRNA), GSE68316 (lncRNA/mRNA) and GSE32453 (mRNA) expression profiles downloaded from the Gene Expression Omnibus datasets. Bioinformatics tools were employed to perform function and pathway enrichment analysis, protein-protein interaction, lncRNA-miRNA-mRNA and hub gene networks. Subsequently, DEGs were used as targets to predict drugs. The results indicated that a total of 2,234 DElncs (1,120 upregulated and 1,114 downregulated), 5 DEmiRs (2 upregulated and 3 downregulated) and 42 DEGs (35 upregulated and 7 downregulated) were identified in 4 microarray profiles. Gene ontology analysis revealed that DEGs were mainly involved in actin filament and stress fiber formation and in calcium ion binding, whereas Kyoto Encyclopedia of Genes and Genomes pathway analysis identified the hypoxia inducible factor-1, transforming growth factor-β and tumor necrosis factor signaling pathways as the main pathways involved in these processes. The hub genes were screened using cytoHubba. A total of 1,086 lncRNA-miRNA-mRNA interactions including 67 lncRNAs, 5 miRNAs and 25 mRNAs were mined in the present study based on prediction websites. Drug prediction indicated that the targeted drugs mainly included angiotensin converting enzyme inhibitors or β-blockers. A comprehensive bioinformatics analysis of the molecular regulatory lncRNA-miRNA-mRNA network was performed and potential therapeutic applications of drugs were predicted in HCM patients. The data may unravel the future molecular mechanism of HCM.
Collapse
Affiliation(s)
- Jiajianghui Li
- Department of Epidemiology and Biostatistics, School of Public Health, Capital Medical University, Beijing 100069, P.R. China
| | - Zining Wu
- Beijing Laboratory for Cardiovascular Precision Medicine, Beijing Anzhen Hospital, Capital Medical University, Beijing 100069, P.R. China
| | - Deqiang Zheng
- Department of Epidemiology and Biostatistics, School of Public Health, Capital Medical University, Beijing 100069, P.R. China
| | - Yue Sun
- Department of Epidemiology and Biostatistics, School of Public Health, Capital Medical University, Beijing 100069, P.R. China
| | - Sisi Wang
- Department of Epidemiology and Biostatistics, School of Public Health, Capital Medical University, Beijing 100069, P.R. China
| | - Yuxiang Yan
- Department of Epidemiology and Biostatistics, School of Public Health, Capital Medical University, Beijing 100069, P.R. China
| |
Collapse
|
57
|
Zhao TX, Mallat Z. Targeting the Immune System in Atherosclerosis. J Am Coll Cardiol 2019; 73:1691-1706. [DOI: 10.1016/j.jacc.2018.12.083] [Citation(s) in RCA: 77] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/17/2018] [Revised: 12/20/2018] [Accepted: 12/30/2018] [Indexed: 02/08/2023]
|
58
|
Heidari L, Ghaderian SMH, Vakili H, Salmani TA. Promoter methylation and functional variants in arachidonate 5-lipoxygenase and forkhead box protein O1 genes associated with coronary artery disease. J Cell Biochem 2019; 120:12360-12368. [PMID: 30825235 DOI: 10.1002/jcb.28501] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2018] [Revised: 12/10/2018] [Accepted: 12/14/2018] [Indexed: 01/08/2023]
Abstract
Coronary artery disease (CAD) is a multifactorial chronic inflammatory disease, which is the most common form of heart disease. This is one of the main causes of death in the United States. Inflammation is one of the key drivers of atherosclerotic plaque development. Forkhead box protein O1 (FOXO1s) family and 5-lipoxygenase make an important contribution to atherosclerosis. The aim of this study was to investigate the methylation pattern and polymorphism analysis of FOXO1 and arachidonate 5-lipoxygenase (ALOX5) promoter genes. We studied 50 patients with CAD and 50 age- and sex-matched healthy controls by high resolution melt technique. Overall, we found significant differences between patients and controls in terms of the promoter methylation of ALOX5 (P > 0.05). But there was no significant difference in FOXO1 promoter methylation between patient and controls. Single nucleotide polymorphisms genotyping of rs12762303 and rs2297627, in ALOX5 and FOXO1 genes were demonstrated a significant correlation between mutant allele and the risk of CAD, respectively. Furthermore, there were significant associations between CT + CC genotype and ALOX5 expression. Our findings demonstrated functional effects of single nucleotide polymorphisms (SNPs) and DNA methylation in ALOX5 on mentioned genes expression and they resulted in CAD progression.
Collapse
Affiliation(s)
- Laleh Heidari
- Department of Medical Genetics, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Sayyed Mohammad Hossein Ghaderian
- Department of Medical Genetics, Shahid Beheshti University of Medical Sciences, Tehran, Iran.,Urogenital Stem Cell Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Hossein Vakili
- Modarres Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Tayyeb Ali Salmani
- Department of Medical Genetics, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
59
|
Chen W, Sinha B, Li Y, Benowitz L, Chen Q, Zhang Z, Patel NJ, Aziz-Sultan AM, Chiocca AE, Wang X. Monogenic, Polygenic, and MicroRNA Markers for Ischemic Stroke. Mol Neurobiol 2019; 56:1330-1343. [PMID: 29948938 PMCID: PMC7358039 DOI: 10.1007/s12035-018-1055-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2017] [Accepted: 03/29/2018] [Indexed: 02/08/2023]
Abstract
Ischemic stroke (IS) is a leading disease with high mortality and disability, as well as with limited therapeutic window. Biomarkers for earlier diagnosis of IS have long been pursued. Family and twin studies confirm that genetic variations play an important role in IS pathogenesis. Besides DNA mutations found previously by genetic linkage analysis for monogenic IS (Mendelian inheritance), recent studies using genome-wide associated study (GWAS) and microRNA expression profiling have resulted in a large number of DNA and microRNA biomarkers in polygenic IS (sporadic IS), especially in different IS subtypes and imaging phenotypes. The present review summarizes genetic markers discovered by clinical studies and discusses their pathogenic molecular mechanisms involved in developmental or regenerative anomalies of blood vessel walls, neuronal apoptosis, excitotoxic death, inflammation, neurogenesis, and angiogenesis. The possible impact of environment on genetics is addressed as well. We also include a perspective on further studies and clinical application of these IS biomarkers.
Collapse
Affiliation(s)
- Wu Chen
- Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA.
- Department of Clinical Laboratory, Dongfeng Hospital, Hubei University of Medicine, Shiyan, 442008, Hubei, China.
| | - Bharati Sinha
- Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
- Department of Newborn Medicine, Brigham and Women's Hospital, Boston, MA, 02115, USA
| | - Yi Li
- Department of Clinical Laboratory, Dongfeng Hospital, Hubei University of Medicine, Shiyan, 442008, Hubei, China
| | - Larry Benowitz
- Department of Neurosurgery, Boston Children's Hospital, F.M. Kirby Neurobiology Center for Life Science, Harvard Medical School, Boston, MA, 02115, USA
| | - Qinhua Chen
- Experimental Center, Dongfeng Hospital, Hubei University of Medicine, Shiyan, 442008, Hubei, China
| | - Zhenghong Zhang
- Department of Neurology, Dongfeng Hospital, Hubei University of Medicine, Shiyan, 442008, Hubei, China
| | - Nirav J Patel
- Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Ali M Aziz-Sultan
- Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Antonio E Chiocca
- Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Xin Wang
- Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA.
| |
Collapse
|
60
|
Zheng JH, Ning GL, Xu WH, Wu XC, Ma XC. Lack of association between ALOX5AP genetic polymorphisms and risk of ischemic stroke: evidence from meta-analyses. Neuropsychiatr Dis Treat 2019; 15:357-367. [PMID: 30774347 PMCID: PMC6354695 DOI: 10.2147/ndt.s182674] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
BACKGROUND In recent years, there has been substantial research evaluating the relationship between arachidonate 5-lipoxygenase-activating protein (ALOX5AP) polymorphisms and ischemic stroke (IS). The objective of this study was to systematically review and analyze the existing evidence. METHODS A comprehensive search of major electronic databases for studies published between 1990 and 2018 was carried out. Data were synthesized as OR and 95% CI using fixed-effects and random-effects models. RESULTS A total of 30 studies were available for analysis. The aggregate sample size across all studies was 32,782 (16,294 cases and 16,488 controls). We found no association of the ALOX5AP rs10507391 (OR=1.03 for A allele vs T allele; 95% CI: 0.93-1.14; P=0.557), rs4769874 (OR=1.13 for A allele vs G allele; 95% CI: 1.00-1.28; P=0.050), rs9551963 (OR=1.03 for A allele vs C allele; 95% CI: 0.96-1.11; P=0.372), rs17222814 (OR=1.09 for A allele vs G allele; 95% CI: 0.96-1.24; P=0.195), rs17222919 (OR=0.89 for G allele vs T allele; 95% CI: 0.75-1.06; P=0.175), and rs4073259 (OR=1.20 for A allele vs G allele; 95% CI: 1.00-1.45; P=0.056) polymorphisms with IS risk. Haplotype analysis also did not yield significant findings for the HapA (rs17222814G-rs10507391T-rs4769874G-rs9551963A; OR=1.20; 95% CI: 0.91-1.56; P=0.192) and HapB (rs17216473A-rs10507391A-rs9315050A-rs17222842G; OR=1.11; 95% CI: 0.90-1.38; P=0.339) haplotypes. CONCLUSION Current evidence does not support an association of rs10507391, rs4769874, rs9551963, rs17222814, rs17222919, rs4073259, and HapA and HapB with IS risk.
Collapse
Affiliation(s)
- Jing-Hui Zheng
- Department of Cardiology, Ruikang Hospital Affiliated to Guangxi University of Chinese Medicine, Nanning 530011, China,
| | - Gui-Lan Ning
- Department of Cardiology, Ruikang Hospital Affiliated to Guangxi University of Chinese Medicine, Nanning 530011, China,
| | - Wen-Hua Xu
- Department of Cardiology, Ruikang Hospital Affiliated to Guangxi University of Chinese Medicine, Nanning 530011, China,
| | - Xin-Cheng Wu
- Department of Cardiology, Ruikang Hospital Affiliated to Guangxi University of Chinese Medicine, Nanning 530011, China,
| | - Xiao-Cong Ma
- Department of Cardiology, Ruikang Hospital Affiliated to Guangxi University of Chinese Medicine, Nanning 530011, China,
| |
Collapse
|
61
|
Kuhnert R, Sárosi MB, George S, Lönnecke P, Hofmann B, Steinhilber D, Steinmann S, Schneider-Stock R, Murganić B, Mijatović S, Maksimović-Ivanić D, Hey-Hawkins E. Carborane-Based Analogues of 5-Lipoxygenase Inhibitors Co-inhibit Heat Shock Protein 90 in HCT116 Cells. ChemMedChem 2018; 14:255-261. [DOI: 10.1002/cmdc.201800651] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2018] [Revised: 11/12/2018] [Indexed: 12/31/2022]
Affiliation(s)
- Robert Kuhnert
- Faculty of Chemistry and Mineralogy; Institute of Inorganic Chemistry; Universität Leipzig; Johannisallee 29 04103 Leipzig Germany
| | - Menyhárt-Botond Sárosi
- Faculty of Chemistry and Mineralogy; Institute of Inorganic Chemistry; Universität Leipzig; Johannisallee 29 04103 Leipzig Germany
| | - Sven George
- Institut für Pharmazeutische Chemie; Johann-Wolfgang-Goethe-Universität Frankfurt; Max-von-Laue-Straße 9 60438 Frankfurt Germany
| | - Peter Lönnecke
- Faculty of Chemistry and Mineralogy; Institute of Inorganic Chemistry; Universität Leipzig; Johannisallee 29 04103 Leipzig Germany
| | - Bettina Hofmann
- Institut für Pharmazeutische Chemie; Johann-Wolfgang-Goethe-Universität Frankfurt; Max-von-Laue-Straße 9 60438 Frankfurt Germany
| | - Dieter Steinhilber
- Institut für Pharmazeutische Chemie; Johann-Wolfgang-Goethe-Universität Frankfurt; Max-von-Laue-Straße 9 60438 Frankfurt Germany
| | - Sara Steinmann
- Pathologisches Institut; Universität Erlangen; Universitätsstraße 22 91054 Erlangen Germany
| | - Regine Schneider-Stock
- Pathologisches Institut; Universität Erlangen; Universitätsstraße 22 91054 Erlangen Germany
| | - Blagoje Murganić
- Department of Immunology, Institute for Biological Research “Sinisa Stankovic”; University of Belgrade; Bul. despota Stefana 142 11060 Belgrade Serbia
| | - Sanja Mijatović
- Department of Immunology, Institute for Biological Research “Sinisa Stankovic”; University of Belgrade; Bul. despota Stefana 142 11060 Belgrade Serbia
| | - Danijela Maksimović-Ivanić
- Department of Immunology, Institute for Biological Research “Sinisa Stankovic”; University of Belgrade; Bul. despota Stefana 142 11060 Belgrade Serbia
| | - Evamarie Hey-Hawkins
- Faculty of Chemistry and Mineralogy; Institute of Inorganic Chemistry; Universität Leipzig; Johannisallee 29 04103 Leipzig Germany
| |
Collapse
|
62
|
Sonnweber T, Pizzini A, Nairz M, Weiss G, Tancevski I. Arachidonic Acid Metabolites in Cardiovascular and Metabolic Diseases. Int J Mol Sci 2018; 19:ijms19113285. [PMID: 30360467 PMCID: PMC6274989 DOI: 10.3390/ijms19113285] [Citation(s) in RCA: 257] [Impact Index Per Article: 42.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2018] [Revised: 10/20/2018] [Accepted: 10/21/2018] [Indexed: 12/20/2022] Open
Abstract
Lipid and immune pathways are crucial in the pathophysiology of metabolic and cardiovascular disease. Arachidonic acid (AA) and its derivatives link nutrient metabolism to immunity and inflammation, thus holding a key role in the emergence and progression of frequent diseases such as obesity, diabetes, non-alcoholic fatty liver disease, and cardiovascular disease. We herein present a synopsis of AA metabolism in human health, tissue homeostasis, and immunity, and explore the role of the AA metabolome in diverse pathophysiological conditions and diseases.
Collapse
Affiliation(s)
- Thomas Sonnweber
- Department of Internal Medicine II, Medical University Innsbruck, Innsbruck 6020, Austria.
| | - Alex Pizzini
- Department of Internal Medicine II, Medical University Innsbruck, Innsbruck 6020, Austria.
| | - Manfred Nairz
- Department of Internal Medicine II, Medical University Innsbruck, Innsbruck 6020, Austria.
| | - Günter Weiss
- Department of Internal Medicine II, Medical University Innsbruck, Innsbruck 6020, Austria.
| | - Ivan Tancevski
- Department of Internal Medicine II, Medical University Innsbruck, Innsbruck 6020, Austria.
| |
Collapse
|
63
|
Ye H, Zhang X, Chen Z, Li X, Zhang T, Yang C, Huang L. Association between the polymorphism (rs17222919, -1316T/G) of 5-lipoxygenase-activating protein gene (ALOX5AP) and the risk of stroke: A meta analysis. Medicine (Baltimore) 2018; 97:e12682. [PMID: 30313062 PMCID: PMC6203560 DOI: 10.1097/md.0000000000012682] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
Abstract
BACKGROUND The objective of this study was to evalutate the relationship between 5-lipoxygenase-activating protein gene (ALOX5AP) -rs17222919-1316T/G polymorphisms and the risk of stroke. METHODS Relative studies were searched in January 2018. Case-control studies with extractable data were selected. Four gene models were analyzed including, allele genetic model (G vs T), additive genetic model (GG vs TT, GT vs TT), recessive genetic model (GG vs GT + TT), and dominant genetic model (GG + GT vs TT). Effect sizes included odds ratio (OR) and 95% confidence interval (CI). Heterogeneity was assessed by using Q test and I test. Publication bias was evaluated by using Egger method. The reliability of the results was assessed with sensitivity analysis. All the data analysis was performed with R 3.10 software. RESULTS A total of 5 studies inclusing 8492 patients were included. There were significant relationship between ALOX5AP-rs17222919-1316T/G polymorphisms and stroke under all models (P < .05) except the additive genetic model GT versus TT (P > .05). No publication bias was noted. Sensitivity analysis indicated that the results were not stable. CONCLUSION This meta-analysis indicates that ALOX5AP-rs17222919-1316T/G may be a protective factor aginst stroke.
Collapse
|
64
|
Araújo AC, Wheelock CE, Haeggström JZ. The Eicosanoids, Redox-Regulated Lipid Mediators in Immunometabolic Disorders. Antioxid Redox Signal 2018; 29:275-296. [PMID: 28978222 DOI: 10.1089/ars.2017.7332] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
SIGNIFICANCE The oxidation of arachidonic acid via cyclooxygenase (COX) and lipoxygenase (LOX) activity to produce eicosanoids during inflammation is a well-known biosynthetic pathway. These lipid mediators are involved in fever, pain, and thrombosis and are produced from multiple cells as well as cell/cell interactions, for example, immune cells and epithelial/endothelial cells. Metabolic disorders, including hyperlipidemia, hypertension, and diabetes, are linked with chronic low-grade inflammation, impacting the immune system and promoting a variety of chronic diseases. Recent Advances: Multiple studies have corroborated the important function of eicosanoids and their receptors in (non)-inflammatory cells in immunometabolic disorders (e.g., insulin resistance, obesity, and cardiovascular and nonalcoholic fatty liver diseases). In this context, LOX and COX products are involved in both pro- and anti-inflammatory responses. In addition, recent work has elucidated the potent function of specialized proresolving mediators (i.e., lipoxins and resolvins) in resolving inflammation, protecting organs, and stimulating tissue repair and remodeling. CRITICAL ISSUES Inhibiting/stimulating selected eicosanoid pathways may result in anti-inflammatory and proresolution responses leading to multiple beneficial effects, including the abrogation of reactive oxygen species production, increased speed of resolution, and overall improvement of diseases related to immunometabolic perturbations. FUTURE DIRECTIONS Despite many achievements, it is crucial to understand the molecular and cellular mechanisms underlying immunological/metabolic cross talk to offer substantial therapeutic promise. Antioxid. Redox Signal. 29, 275-296.
Collapse
Affiliation(s)
- Ana Carolina Araújo
- Division of Physiological Chemistry II, Department of Medical Biochemistry and Biophysics, Karolinska Institutet , Stockholm, Sweden
| | - Craig E Wheelock
- Division of Physiological Chemistry II, Department of Medical Biochemistry and Biophysics, Karolinska Institutet , Stockholm, Sweden
| | - Jesper Z Haeggström
- Division of Physiological Chemistry II, Department of Medical Biochemistry and Biophysics, Karolinska Institutet , Stockholm, Sweden
| |
Collapse
|
65
|
Tsekmekidou XA, Kotsa KD, Tsetsos FS, Didangelos TP, Georgitsi MA, Roumeliotis AK, Panagoutsos SA, Thodis ED, Theodoridis MT, Papanas NP, Papazoglou DA, Pasadakis PS, Eustratios MS, Paschou PI, Yovos JG. Assessment of association between lipoxygenase genes variants in elderly Greek population and type 2 diabetes mellitus. Diab Vasc Dis Res 2018; 15:340-343. [PMID: 29392977 DOI: 10.1177/1479164118756241] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
BACKGROUND Inflammation plays a pivotal role in the pathogenesis of diabetes and its complications. Arachidonic acid lipoxygenases have been intensively studied in their role in inflammation in metabolic pathways. Thus, we aimed to explore variants of lipoxygenase genes (arachidonate lipoxygenase genes) in a diabetes adult population using a case-control study design. METHODS Study population consisted of 1285 elderly participants, 716 of whom had type 2 diabetes mellitus. The control group consisted of non-diabetes individuals with no history of diabetes history and with a glycated haemoglobin <6.5% (<48 mmol/mol)] and fasting plasma glucose levels <126 mg/dL. Blood samples were genotyped on Illumina Infinium PsychArray. Variants of ALOX5, ALOX5AP, ALOX12, ALOX15 were selected. All statistical analyses were undertaken within PLINK and SPSS packages utilising permutation analysis tests. RESULTS Our findings showed an association of rs9669952 (odds ratio = 0.738, p = 0.013) and rs1132340 (odds ratio = 0.652, p = 0.008) in ALOX5AP and rs11239524 in ALOX5 gene with disease (odds ratio = 0.808, p = 0.038). Rs9315029 which is located near arachidonate ALOX5AP also associated with type 2 diabetes mellitus ( p = 0.025). No variant of ALOX12 and ALOX15 genes associated with disease. CONCLUSION These results indicate a potential protective role of ALOX5AP and 5-arachidonate lipoxygenase gene in diabetes pathogenesis, indicating further the importance of the relationship between diabetes and inflammation. Larger population studies are required to replicate our findings.
Collapse
Affiliation(s)
- Xanthippi A Tsekmekidou
- 1 Division of Endocrinology and Metabolism-Diabetes Center, 1st Internal Medicine Department, Medical School, AHEPA University Hospital, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Kalliopi D Kotsa
- 1 Division of Endocrinology and Metabolism-Diabetes Center, 1st Internal Medicine Department, Medical School, AHEPA University Hospital, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Fotis S Tsetsos
- 2 Department of Molecular Biology and Genetics, Democritus University of Thrace, Alexandroupolis, Greece
| | - Triantafyllos P Didangelos
- 3 Diabetes Center, 1st Propedeutic Internal Medicine Department, AHEPA University Hospital, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Marianthi A Georgitsi
- 2 Department of Molecular Biology and Genetics, Democritus University of Thrace, Alexandroupolis, Greece
- 4 Laboratory of General Biology-Genetics, Department of Medicine, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | | | | | - Elias D Thodis
- 5 Department of Nephrology, Democritus University of Thrace, Alexandroupolis, Greece
| | - Marios T Theodoridis
- 5 Department of Nephrology, Democritus University of Thrace, Alexandroupolis, Greece
| | - Nikolaos P Papanas
- 6 Diabetes Centre, Second Department of Internal Medicine, Democritus University of Thrace, Alexandroupolis, Greece
| | - Dimitrios A Papazoglou
- 6 Diabetes Centre, Second Department of Internal Medicine, Democritus University of Thrace, Alexandroupolis, Greece
| | - Ploumis S Pasadakis
- 5 Department of Nephrology, Democritus University of Thrace, Alexandroupolis, Greece
| | - Maltezos S Eustratios
- 6 Diabetes Centre, Second Department of Internal Medicine, Democritus University of Thrace, Alexandroupolis, Greece
| | - Peristera I Paschou
- 2 Department of Molecular Biology and Genetics, Democritus University of Thrace, Alexandroupolis, Greece
| | - John G Yovos
- 1 Division of Endocrinology and Metabolism-Diabetes Center, 1st Internal Medicine Department, Medical School, AHEPA University Hospital, Aristotle University of Thessaloniki, Thessaloniki, Greece
| |
Collapse
|
66
|
Mawhin MA, Tilly P, Zirka G, Charles AL, Slimani F, Vonesch JL, Michel JB, Bäck M, Norel X, Fabre JE. Neutrophils recruited by leukotriene B4 induce features of plaque destabilization during endotoxaemia. Cardiovasc Res 2018; 114:1656-1666. [DOI: 10.1093/cvr/cvy130] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/04/2018] [Accepted: 05/17/2018] [Indexed: 12/22/2022] Open
Affiliation(s)
- Marie-Anne Mawhin
- UMR 1148 INSERM, Xavier Bichat Hospital, 46 rue Henri Huchard, Paris, France
- IGMBC, Illkirch, France
- UMR 7104 CNRS, Illkirch, France
- U964 INSERM, Illkirch, France
- Strasbourg University, Strasbourg, France
| | - Peggy Tilly
- IGMBC, Illkirch, France
- UMR 7104 CNRS, Illkirch, France
- U964 INSERM, Illkirch, France
- Strasbourg University, Strasbourg, France
| | - Gaia Zirka
- UMR 1148 INSERM, Xavier Bichat Hospital, 46 rue Henri Huchard, Paris, France
| | - Anne-Laure Charles
- Equipe d'accueil 3072, Faculty of Medicine, Translational Medicine Federation, Strasbourg University, Strasbourg, France
| | - Farid Slimani
- IGMBC, Illkirch, France
- UMR 7104 CNRS, Illkirch, France
- U964 INSERM, Illkirch, France
- Strasbourg University, Strasbourg, France
| | | | | | - Magnus Bäck
- Department of Medicine, Karolinska Institutet, Stockholm, Sweden
- INSERM U1116, University of Lorraine and CHRU, Nancy, France
| | - Xavier Norel
- UMR 1148 INSERM, Xavier Bichat Hospital, 46 rue Henri Huchard, Paris, France
| | - Jean-Etienne Fabre
- UMR 1148 INSERM, Xavier Bichat Hospital, 46 rue Henri Huchard, Paris, France
| |
Collapse
|
67
|
Iribarren C, Lu M, Jorgenson E, Martínez M, Lluis-Ganella C, Subirana I, Salas E, Elosua R. Weighted Multi-marker Genetic Risk Scores for Incident Coronary Heart Disease among Individuals of African, Latino and East-Asian Ancestry. Sci Rep 2018; 8:6853. [PMID: 29717161 PMCID: PMC5931622 DOI: 10.1038/s41598-018-25128-x] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2018] [Accepted: 03/22/2018] [Indexed: 02/06/2023] Open
Abstract
We examined the clinical utility of two multi-locus genetic risk scores (GRSs) previously validated in Europeans among persons of African (AFR; n = 2,089), Latino (LAT; n = 4,349) and East-Asian (EA; n = 4,804) ancestry. We used data from the GERA cohort (30-79 years old, 68 to 73% female). We utilized two GRSs with 12 and 51 SNPs, respectively, and the Framingham Risk Score (FRS) to estimate 10-year CHD risk. After a median 8.7 years of follow-up, 450 incident CHD events were documented (95 in AFR, 316 in LAT and 39 EA, respectively). In a model adjusting for principal components and risk factors, tertile 3 vs. tertile 1 of GRS_12 was associated with 1.86 (95% CI, 1.15-3.01), 1.52 (95% CI, 1.02-2.25) and 1.19 (95% CI, 0.77-1.83) increased hazard of CHD in AFR, LAT and EA, respectively. Inclusion of the GRSs in models containing the FRS did not increase the C-statistic but resulted in net overall reclassification of 10% of AFR, 7% LAT and EA and in reclassification of 13% of AFR and EA as well as 10% LAT in the intermediate FRS risk subset. Our results support the usefulness of incorporating genetic information into risk assessment for primary prevention among minority subjects in the U.S.
Collapse
Affiliation(s)
- Carlos Iribarren
- Kaiser Permanente Northern California Division of Research, Oakland, CA, USA.
| | - Meng Lu
- Kaiser Permanente Northern California Division of Research, Oakland, CA, USA
| | - Eric Jorgenson
- Kaiser Permanente Northern California Division of Research, Oakland, CA, USA
| | | | | | - Isaac Subirana
- CIBER of Epidemiology and Public Health, Barcelona, Spain.,Cardiovascular Epidemiology and Genetics, IMIM, Barcelona, Spain
| | | | - Roberto Elosua
- Cardiovascular Epidemiology and Genetics, IMIM, Barcelona, Spain.,CIBER of Cardiovascular Diseases (CIBERCV), Barcelona, Spain
| |
Collapse
|
68
|
Gao X, Sun Q, Zhang W, Jiang Y, Li R, Ye J. Anti-inflammatory effect and mechanism of the spirocyclopiperazinium salt compound LXM-15 in rats and mice. Inflamm Res 2018; 67:363-370. [PMID: 29302720 DOI: 10.1007/s00011-017-1127-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2017] [Revised: 12/05/2017] [Accepted: 12/28/2017] [Indexed: 01/14/2023] Open
Abstract
OBJECTIVE This study aimed to investigate the anti-inflammatory effects of a novel spirocyclopiperazinium salt compound LXM-15, and explore the underlying mechanisms. METHODS Xylene-induced mouse ear oedema and carrageenan-induced rat paw oedema tests were used to evaluate the anti-inflammatory effects of LXM-15. The protein levels of TNF-α, IL-6, phosphorylation of Janus kinase 2 (JAK2) and signal transducer and activator of transcription 3 (STAT3) were detected by ELISA or Western blot analysis. Additionally, receptor blocking test was performed to explore the possible target. RESULTS Intragastric administration with LXM-15 (2, 1, 0.5 mg/kg in mice, and 6, 3, 1.5 mg/kg in rats) produced distinct anti-inflammatory effects in vivo, the highest inhibition percentage was 60 and 52%, respectively (P < 0.01). Following treatment with LXM-15 (6 mg/kg, i.g.), the levels of TNF-α and IL-6 in the rats paws were attenuated by 40 and 41%; and the phosphorylation of JAK2 and STAT3 was restrained by 35 and 45%, respectively (P < 0.01). All effects of LXM-15 were blocked by pretreatment with methyllycaconitine citrate or tropicamide. CONCLUSION This study provides the first report that the spirocyclopiperazinium salt compound LXM-15 displays considerable anti-inflammatory effects. The underlying mechanism may be through activating the peripheral α7 nicotinic acetylcholine receptor and M4 muscarinic acetylcholine receptor, leading to the inhibition of the JAK2/STAT3 signalling pathway, eventually resulting in the reduction of TNF-α and IL-6.
Collapse
Affiliation(s)
- Xiaoli Gao
- Department of Molecular and Cellular Pharmacology, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, People's Republic of China
- Modern Research Center for Traditional Chinese Medicine, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 100029, People's Republic of China
| | - Qi Sun
- Department of Chemical Biology, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, People's Republic of China
| | - Weiwei Zhang
- Department of Molecular and Cellular Pharmacology, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, People's Republic of China
- Clinical Pharmacy of Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, Beijing, 102218, People's Republic of China
| | - Yimin Jiang
- Centre of Medical and Health Analysis, Peking University, Beijing, 100191, People's Republic of China
| | - Runtao Li
- Department of Chemical Biology, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, People's Republic of China
| | - Jia Ye
- Department of Molecular and Cellular Pharmacology, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, People's Republic of China.
| |
Collapse
|
69
|
Ericsson H, Nelander K, Lagerstrom-Fermer M, Balendran C, Bhat M, Chialda L, Gan LM, Heijer M, Kjaer M, Lambert J, Lindstedt EL, Forsberg GB, Whatling C, Skrtic S. Initial Clinical Experience with AZD5718, a Novel Once Daily Oral 5-Lipoxygenase Activating Protein Inhibitor. Clin Transl Sci 2018. [PMID: 29517132 PMCID: PMC5944575 DOI: 10.1111/cts.12546] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
We evaluated safety, tolerability, pharmacokinetics, and pharmacodynamics of AZD5718, a novel 5-lipooxygenase activating protein (FLAP) inhibitor, in a randomized, single-blind, placebo-controlled, first-in-human (FIH) study consisting of single and multiple ascending dosing (SAD and MAD) for 10 days in healthy subjects. Target engagement was measured by ex vivo calcium ionophore stimulated leukotriene B (LTB4 ) production in whole blood and endogenous leukotriene E (LTE4 ) in urine. No clinically relevant safety and tolerability findings were observed. The AZD5718 was rapidly absorbed and plasma concentrations declined biphasically with a mean terminal half-life of 10-12 h. Steady-state levels were achieved after ∼3 days. After both SADs and MADs, a dose/concentration-effect relationship between both LTB4 and LTE4 vs. AZD5718 exposure was observed with concentration of half inhibition (IC50 ) values in the lower nM range. Based on obtained result, AZD5718 is considered as a suitable drug candidate for future evaluation in patients with coronary artery disease (CAD).
Collapse
Affiliation(s)
- Hans Ericsson
- Early Clinical Development, AstraZeneca, Gothenburg, Sweden
| | - Karin Nelander
- Early Clinical Development, AstraZeneca, Gothenburg, Sweden
| | | | - Clare Balendran
- Precision Medicine and Genomics, IMED Biotech Unit, AstraZeneca Gothenburg, Sweden
| | - Maria Bhat
- Precision Medicine and Genomics, IMED Biotech Unit, AstraZeneca Gothenburg, Sweden
| | - Ligia Chialda
- PAREXEL International, Northwick Park Hospital, Harrow, UK
| | - Li-Ming Gan
- Early Clinical Development, AstraZeneca, Gothenburg, Sweden
| | - Maria Heijer
- Early Clinical Development, AstraZeneca, Gothenburg, Sweden
| | - Magnus Kjaer
- Early Clinical Development, AstraZeneca, Gothenburg, Sweden
| | - John Lambert
- PAREXEL International, Northwick Park Hospital, Harrow, UK
| | | | | | - Carl Whatling
- Cardiovascular and Metabolic Diseases, AstraZeneca, Gothenburg, Sweden
| | - Stanko Skrtic
- Early Clinical Development, AstraZeneca, Gothenburg, Sweden
| |
Collapse
|
70
|
Hensley MK, Levine A, Gladwin MT, Lai YC. Emerging therapeutics in pulmonary hypertension. Am J Physiol Lung Cell Mol Physiol 2018; 314:L769-L781. [PMID: 29388467 DOI: 10.1152/ajplung.00259.2017] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Pulmonary hypertension (PH) is a progressive and often fatal illness presenting with nonspecific symptoms of dyspnea, lower extremity edema, and exercise intolerance. Pathologically, endothelial dysfunction leads to abnormal intimal and smooth muscle proliferation along with reduced apoptosis, resulting in increased pulmonary vascular resistance and elevated pulmonary pressures. PH is subdivided into five World Health Organization groups based on the disease pathology and specific cause. While there are Food and Drug Administration-approved medications for the treatment of pulmonary arterial hypertension (PAH; Group 1 PH), as well as for chronic thromboembolic PH (Group 4 PH), the morbidity and mortality remain high. Moreover, there are no approved therapies for other forms of PH (Groups 2, 3, and 5) at present. New research has identified molecular targets that mediate vasodilation, anti-inflammatory, and antifibrotic changes within the pulmonary vasculature. Given that PAH is the most commonly studied form of PH worldwide and because recent studies have led to better mechanistic understanding of this devastating disease, in this review we attempt to provide an updated overview of new therapeutic approaches under investigation for the treatment of PH, with a particular focus on PAH, as well as to offer guidelines for future investigations.
Collapse
Affiliation(s)
- Matthew K Hensley
- Division of Pulmonary and Critical Care Medicine, University of Michigan , Ann Arbor, Michigan
| | - Andrea Levine
- Pittsburgh Heart, Lung, Blood and Vascular Medicine Institute, University of Pittsburgh , Pittsburgh, Pennsylvania.,Division of Pulmonary, Allergy and Critical Care Medicine, University of Pittsburgh School of Medicine , Pittsburgh, Pennsylvania
| | - Mark T Gladwin
- Pittsburgh Heart, Lung, Blood and Vascular Medicine Institute, University of Pittsburgh , Pittsburgh, Pennsylvania.,Division of Pulmonary, Allergy and Critical Care Medicine, University of Pittsburgh School of Medicine , Pittsburgh, Pennsylvania
| | - Yen-Chun Lai
- Pittsburgh Heart, Lung, Blood and Vascular Medicine Institute, University of Pittsburgh , Pittsburgh, Pennsylvania.,Division of Pulmonary, Allergy and Critical Care Medicine, University of Pittsburgh School of Medicine , Pittsburgh, Pennsylvania
| |
Collapse
|
71
|
Yurdagul A, Doran AC, Cai B, Fredman G, Tabas IA. Mechanisms and Consequences of Defective Efferocytosis in Atherosclerosis. Front Cardiovasc Med 2018. [PMID: 29379788 DOI: 10.3389/fcvm.2017.00086e] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Efficient clearance of apoptotic cells, termed efferocytosis, critically regulates normal homeostasis whereas defective uptake of apoptotic cells results in chronic and non-resolving inflammatory diseases, such as advanced atherosclerosis. Monocyte-derived macrophages recruited into developing atherosclerotic lesions initially display efficient efferocytosis and temper inflammatory responses, processes that restrict plaque progression. However, during the course of plaque development, macrophages undergo cellular reprogramming that reduces efferocytic capacity, which results in post-apoptotic necrosis of apoptotic cells and inflammation. Furthermore, defective efferocytosis in advanced atherosclerosis is a major driver of necrotic core formation, which can trigger plaque rupture and acute thrombotic cardiovascular events. In this review, we discuss the molecular and cellular mechanisms that regulate efferocytosis, how efferocytosis promotes the resolution of inflammation, and how defective efferocytosis leads to the formation of clinically dangerous atherosclerotic plaques.
Collapse
Affiliation(s)
- Arif Yurdagul
- Department of Medicine, Columbia University, New York, NY, United States.,Department of Pathology and Cell Biology, Columbia University, New York, NY, United States.,Department of Physiology, Columbia University, New York, NY, United States
| | - Amanda C Doran
- Department of Medicine, Columbia University, New York, NY, United States.,Department of Pathology and Cell Biology, Columbia University, New York, NY, United States.,Department of Physiology, Columbia University, New York, NY, United States
| | - Bishuang Cai
- Department of Medicine, Columbia University, New York, NY, United States.,Department of Pathology and Cell Biology, Columbia University, New York, NY, United States.,Department of Physiology, Columbia University, New York, NY, United States
| | - Gabrielle Fredman
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY, United States
| | - Ira A Tabas
- Department of Medicine, Columbia University, New York, NY, United States.,Department of Pathology and Cell Biology, Columbia University, New York, NY, United States.,Department of Physiology, Columbia University, New York, NY, United States
| |
Collapse
|
72
|
Yurdagul A, Doran AC, Cai B, Fredman G, Tabas IA. Mechanisms and Consequences of Defective Efferocytosis in Atherosclerosis. Front Cardiovasc Med 2018; 4:86. [PMID: 29379788 PMCID: PMC5770804 DOI: 10.3389/fcvm.2017.00086] [Citation(s) in RCA: 167] [Impact Index Per Article: 27.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2017] [Accepted: 12/11/2017] [Indexed: 12/22/2022] Open
Abstract
Efficient clearance of apoptotic cells, termed efferocytosis, critically regulates normal homeostasis whereas defective uptake of apoptotic cells results in chronic and non-resolving inflammatory diseases, such as advanced atherosclerosis. Monocyte-derived macrophages recruited into developing atherosclerotic lesions initially display efficient efferocytosis and temper inflammatory responses, processes that restrict plaque progression. However, during the course of plaque development, macrophages undergo cellular reprogramming that reduces efferocytic capacity, which results in post-apoptotic necrosis of apoptotic cells and inflammation. Furthermore, defective efferocytosis in advanced atherosclerosis is a major driver of necrotic core formation, which can trigger plaque rupture and acute thrombotic cardiovascular events. In this review, we discuss the molecular and cellular mechanisms that regulate efferocytosis, how efferocytosis promotes the resolution of inflammation, and how defective efferocytosis leads to the formation of clinically dangerous atherosclerotic plaques.
Collapse
Affiliation(s)
- Arif Yurdagul
- Department of Medicine, Columbia University, New York, NY, United States.,Department of Pathology and Cell Biology, Columbia University, New York, NY, United States.,Department of Physiology, Columbia University, New York, NY, United States
| | - Amanda C Doran
- Department of Medicine, Columbia University, New York, NY, United States.,Department of Pathology and Cell Biology, Columbia University, New York, NY, United States.,Department of Physiology, Columbia University, New York, NY, United States
| | - Bishuang Cai
- Department of Medicine, Columbia University, New York, NY, United States.,Department of Pathology and Cell Biology, Columbia University, New York, NY, United States.,Department of Physiology, Columbia University, New York, NY, United States
| | - Gabrielle Fredman
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY, United States
| | - Ira A Tabas
- Department of Medicine, Columbia University, New York, NY, United States.,Department of Pathology and Cell Biology, Columbia University, New York, NY, United States.,Department of Physiology, Columbia University, New York, NY, United States
| |
Collapse
|
73
|
Wierer M, Prestel M, Schiller HB, Yan G, Schaab C, Azghandi S, Werner J, Kessler T, Malik R, Murgia M, Aherrahrou Z, Schunkert H, Dichgans M, Mann M. Compartment-resolved Proteomic Analysis of Mouse Aorta during Atherosclerotic Plaque Formation Reveals Osteoclast-specific Protein Expression. Mol Cell Proteomics 2017; 17:321-334. [PMID: 29208753 PMCID: PMC5795394 DOI: 10.1074/mcp.ra117.000315] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2017] [Revised: 11/20/2017] [Indexed: 12/29/2022] Open
Abstract
Atherosclerosis leads to vascular lesions that involve major rearrangements of the vascular proteome, especially of the extracellular matrix (ECM). Using single aortas from ApoE knock out mice, we quantified formation of plaques by single-run, high-resolution mass spectrometry (MS)-based proteomics. To probe localization on a proteome-wide scale we employed quantitative detergent solubility profiling. This compartment- and time-resolved resource of atherogenesis comprised 5117 proteins, 182 of which changed their expression status in response to vessel maturation and atherosclerotic plaque development. In the insoluble ECM proteome, 65 proteins significantly changed, including relevant collagens, matrix metalloproteinases and macrophage derived proteins. Among novel factors in atherosclerosis, we identified matrilin-2, the collagen IV crosslinking enzyme peroxidasin as well as the poorly characterized MAM-domain containing 2 (Mamdc2) protein as being up-regulated in the ECM during atherogenesis. Intriguingly, three subunits of the osteoclast specific V-ATPase complex were strongly increased in mature plaques with an enrichment in macrophages thus implying an active de-mineralization function.
Collapse
Affiliation(s)
- Michael Wierer
- From the ‡Department of Proteomics and Signal Transduction, Max-Planck Institute of Biochemistry, Martinsried, Germany
| | - Matthias Prestel
- §Institute for Stroke and Dementia Research, Klinikum der Universität München, München, Germany
| | - Herbert B Schiller
- From the ‡Department of Proteomics and Signal Transduction, Max-Planck Institute of Biochemistry, Martinsried, Germany.,¶Comprehensive Pneumology Center, Helmholtz Zentrum München, Member of the German Center for Lung Research (DZL), Munich, Germany
| | - Guangyao Yan
- §Institute for Stroke and Dementia Research, Klinikum der Universität München, München, Germany
| | - Christoph Schaab
- From the ‡Department of Proteomics and Signal Transduction, Max-Planck Institute of Biochemistry, Martinsried, Germany
| | - Sepiede Azghandi
- §Institute for Stroke and Dementia Research, Klinikum der Universität München, München, Germany
| | - Julia Werner
- ‖Klinik für Herz- und Kreislauferkrankungen, Deutsches Herzzentrum München, Technische Universität München, Munich, Germany
| | - Thorsten Kessler
- ‖Klinik für Herz- und Kreislauferkrankungen, Deutsches Herzzentrum München, Technische Universität München, Munich, Germany
| | - Rainer Malik
- §Institute for Stroke and Dementia Research, Klinikum der Universität München, München, Germany
| | - Marta Murgia
- From the ‡Department of Proteomics and Signal Transduction, Max-Planck Institute of Biochemistry, Martinsried, Germany.,**Department of Biomedical Sciences, University of Padova, Padua, Italy
| | - Zouhair Aherrahrou
- ‡‡Institut für Integrative und Experimentelle Genomik, Universität zu Lübeck, Lübeck, Germany.,§§Deutsches Zentrum für Herz-Kreislauf-Forschung (DZHK), e.V., Partner Site Hamburg/Kiel/Lübeck, Lübeck Germany
| | - Heribert Schunkert
- ‖Klinik für Herz- und Kreislauferkrankungen, Deutsches Herzzentrum München, Technische Universität München, Munich, Germany.,¶¶DZHK e.V. (German Center for Cardiovascular Research), Partner Site Munich Heart Alliance, Munich, Germany
| | - Martin Dichgans
- §Institute for Stroke and Dementia Research, Klinikum der Universität München, München, Germany;
| | - Matthias Mann
- From the ‡Department of Proteomics and Signal Transduction, Max-Planck Institute of Biochemistry, Martinsried, Germany;
| |
Collapse
|
74
|
Identification of risk genes associated with myocardial infarction based on the recursive feature elimination algorithm and support vector machine classifier. Mol Med Rep 2017; 17:1555-1560. [PMID: 29138828 PMCID: PMC5780094 DOI: 10.3892/mmr.2017.8044] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2017] [Accepted: 10/02/2017] [Indexed: 02/06/2023] Open
Abstract
The aim of the present study was to identify risk genes in myocardial infarction. Microarray data GSE34198, containing data from the peripheral blood of 49 myocardial infarction samples and 48 corresponding control samples, were downloaded from the Gene Expression Omnibus database to screen the differentially expressed genes (DEGs). The DEGs were used to construct a protein-protein interaction (PPI) network of patient samples, from which the feature genes were identified using the neighboring score method. The recursive feature elimination (RFE) algorithm was employed to select the risk genes among feature genes, which were subsequently applied to perform a support vector machine (SVM) classifier to identify the specific signature in myocardial infarction samples. Another dataset, GSE61144, was also downloaded to verify the efficacy of the classifier. A total of 724 downregulated and 483 upregulated DEGs were screened in patient samples compared with control samples in the GSE34198 dataset. The PPI network of myocardial infarction was comprised of 1,083 nodes (genes) and 46,363 lines (connections). Using the neighborhood scoring method, the top 100 feature genes in myocardial infarction samples were identified as the disease feature genes, which distinguish the myocardial infarction samples from the control samples. The RFE algorithm screened 15 risk genes, which were employed to construct a SVM classifier with an average precision of 88% to the patient sample following visualization by a confusion matrix. The predictive precision of the classifier on another microarray dataset, GSE61144, was 0.92, with an average true positive of 0.9278 and an average false positive of 0.2361. A-kinase-anchoring protein 12 (AKAP12) and glycine receptor α2 (GLRA2) were two risk genes in the SVM classifier. Therefore, AKAP12 and GLRA2 exert potential roles in the development of myocardial infarction, potentially by influencing cardiac contractility and protecting against ischemia-reperfusion injury, which may provide clues in developing potential diagnostic biomarkers or therapeutic targets for myocardial infarction.
Collapse
|
75
|
García-Heredia JM, Carnero A. The cargo protein MAP17 (PDZK1IP1) regulates the immune microenvironment. Oncotarget 2017; 8:98580-98597. [PMID: 29228712 PMCID: PMC5716752 DOI: 10.18632/oncotarget.21651] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2017] [Accepted: 08/25/2017] [Indexed: 02/06/2023] Open
Abstract
Inflammation is a complex defensive response activated after various harmful stimuli allowing the clearance of damaged cells and initiating healing and regenerative processes. Chronic, or pathological, inflammation is also one of the causes of neoplastic transformation and cancer development. MAP17 is a cargo protein that transports membrane proteins from the endoplasmic reticulum. Therefore, its overexpression may be linked to an excess of membrane proteins that may be recognized as an unwanted signal, triggering local inflammation. Therefore, we analyzed whether its overexpression is related to an inflammatory phenotype. In this work, we found a correlation between MAP17 expression and inflammatory phenotype in tumors and in other inflammatory diseases such as Crohn's disease, Barrett's esophagus, COPD or psoriasis. MAP17 expression correlated also with the markers of inflammation HLAs, BBS10, HERC2, ADNP and PYCARD. Furthermore, we found that MAP17 expression directly regulates NFAT2 and IL-6 activation, inducing the differentiation of monocytes to dendritic cells and suggesting a causal role of MAP17 in inflammation. Immunohistochemistry confirms local inflammation, mainly CD45+ cells, at the site of expression of MAP17, at least in tumors, Crohn's and psoriasis. Therefore, our data indicates that the overexpression of the protein MAP17 plays important role in diseases involving chronic inflammation.
Collapse
Affiliation(s)
- José M García-Heredia
- Instituto de Biomedicina de Sevilla, IBIS/Hospital Universitario Virgen del Rocío/Universidad de Sevilla/Consejo Superior de Investigaciones Científicas, Seville, Spain.,Department of Vegetal Biochemistry and Molecular Biology, University of Seville, Seville, Spain.,CIBER de Cáncer, Instituto de Salud Carlos III, Madrid, Spain
| | - Amancio Carnero
- Instituto de Biomedicina de Sevilla, IBIS/Hospital Universitario Virgen del Rocío/Universidad de Sevilla/Consejo Superior de Investigaciones Científicas, Seville, Spain.,CIBER de Cáncer, Instituto de Salud Carlos III, Madrid, Spain
| |
Collapse
|
76
|
Genética de la cardiopatía isquémica: del conocimiento actual a las implicaciones clínicas. Rev Esp Cardiol 2017. [DOI: 10.1016/j.recesp.2017.02.033] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
|
77
|
Role of the Cysteinyl Leukotrienes in the Pathogenesis and Progression of Cardiovascular Diseases. Mediators Inflamm 2017; 2017:2432958. [PMID: 28932020 PMCID: PMC5592403 DOI: 10.1155/2017/2432958] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2017] [Accepted: 08/17/2017] [Indexed: 12/18/2022] Open
Abstract
Cysteinyl leukotrienes (CysLTs) are potent lipid inflammatory mediators synthesized from arachidonic acid, through the 5-lipoxygenase (5-LO) pathway. Owing to their properties, CysLTs play a crucial role in the pathogenesis of inflammation; therefore, CysLT modifiers as synthesis inhibitors or receptor antagonists, central in asthma management, may become a potential target for the treatment of other inflammatory diseases such as the cardiovascular disorders. 5-LO pathway activation and increased expression of its mediators and receptors are found in cardiovascular diseases. Moreover, the cardioprotective effects observed by using CysLT modifiers are promising and contribute to elucidate the link between CysLTs and cardiovascular disease. The aim of this review is to summarize the state of present research about the role of the CysLTs in the pathogenesis and progression of atherosclerosis and myocardial infarction.
Collapse
|
78
|
Fredman G, Tabas I. Boosting Inflammation Resolution in Atherosclerosis: The Next Frontier for Therapy. THE AMERICAN JOURNAL OF PATHOLOGY 2017; 187:1211-1221. [PMID: 28527709 DOI: 10.1016/j.ajpath.2017.01.018] [Citation(s) in RCA: 133] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 01/23/2017] [Accepted: 01/30/2017] [Indexed: 02/08/2023]
Abstract
Defective inflammation resolution is the underlying cause of prevalent chronic inflammatory diseases, such as arthritis, asthma, cancer, and neurodegenerative and cardiovascular diseases. Inflammation resolution is governed by several endogenous factors, including fatty acid-derived specialized proresolving mediators and proteins, such as annexin A1. Specifically, specialized proresolving mediators comprise a family of mediators that include arachidonic acid-derived lipoxins, omega-3 fatty acid eicosapentaenoic acid-derived resolvins, docosahexaenoic acid-derived resolvins, protectins, and maresins. Emerging evidence indicates that imbalances between specialized proresolving mediators and proinflammatory mediators are associated with several prevalent human diseases, including atherosclerosis. Mechanisms that drive this imbalance remain largely unknown and will be discussed in this review. Furthermore, the concept of dysregulated inflammation resolution in atherosclerosis has been known for several decades. Recently, there has been an explosion of new work with regard to the therapeutic application of proresolving ligands in experimental atherosclerosis. Therefore, this review will highlight recent advances in our understanding of how inflammation resolution may become defective in atherosclerosis and the potential for proresolving therapeutics in atherosclerosis. Last, we offer insight for future implications of the field.
Collapse
Affiliation(s)
- Gabrielle Fredman
- Department of Molecular and Cellular Physiology, Center for Cardiovascular Sciences, Albany Medical College, Albany, New York.
| | - Ira Tabas
- Departments of Medicine, Pathology and Cell Biology, and Physiology, Columbia University Medical Center, New York, New York
| |
Collapse
|
79
|
Abstract
Lipids are potent signaling molecules that regulate a multitude of cellular responses, including cell growth and death and inflammation/infection, via receptor-mediated pathways. Derived from polyunsaturated fatty acids (PUFAs), such as arachidonic acid (AA), eicosapentaenoic acid (EPA), and docosahexaenoic acid (DHA), each lipid displays unique properties, thus making their role in inflammation distinct from that of other lipids derived from the same PUFA. This diversity arises from their synthesis, which occurs via discrete enzymatic pathways and because they elicit responses via different receptors. This review will collate the bioactive lipid research to date and summarize the major pathways involved in their biosynthesis and role in inflammation. Specifically, lipids derived from AA (prostanoids, leukotrienes, 5-oxo-6,8,11,14-eicosatetraenoic acid, lipoxins, and epoxyeicosatrienoic acids), EPA (E-series resolvins), and DHA (D-series resolvins, protectins, and maresins) will be discussed herein.
Collapse
|
80
|
Pace S, Pergola C, Dehm F, Rossi A, Gerstmeier J, Troisi F, Pein H, Schaible AM, Weinigel C, Rummler S, Northoff H, Laufer S, Maier TJ, Rådmark O, Samuelsson B, Koeberle A, Sautebin L, Werz O. Androgen-mediated sex bias impairs efficiency of leukotriene biosynthesis inhibitors in males. J Clin Invest 2017; 127:3167-3176. [PMID: 28737505 DOI: 10.1172/jci92885] [Citation(s) in RCA: 72] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2017] [Accepted: 04/28/2017] [Indexed: 12/13/2022] Open
Abstract
Proinflammatory leukotrienes (LTs) are produced by 5-lipoxygenase (5-LO) aided by 5-LO-activating protein (FLAP). LT biosynthesis inhibitors are currently under clinical investigation as treatments for respiratory and cardiovascular diseases. Here, we have revealed a sex bias in the efficiency of clinically relevant LT biosynthesis inhibitors, showing that their effects are superior in females. We found that androgens cause these sex differences by impeding the LT-biosynthetic 5-LO/FLAP complex assembly. Lower doses of the FLAP inhibitor MK886 were required to reduce LTB4 levels in exudates of female versus male mice and rats. Following platelet-activating factor-induced shock, MK886 increased survival exclusively in female mice, and this effect was abolished by testosterone administration. FLAP inhibitors and the novel-type 5-LO inhibitors licofelone and sulindac sulfide exhibited higher potencies in human blood from females, and bioactive 5-LO/FLAP complexes were formed in female, but not male, human and murine leukocytes. Supplementation of female blood or leukocytes with 5α-dihydrotestosterone abolished the observed sex differences. Our data suggest that females may benefit from anti-LT therapy to a greater extent than males, prompting consideration of sex issues in LT modifier development.
Collapse
Affiliation(s)
- Simona Pace
- Department of Pharmaceutical/Medicinal Chemistry, Institute of Pharmacy, Friedrich-Schiller-University Jena, University Hospital Jena, Jena, Germany
| | - Carlo Pergola
- Department of Pharmaceutical/Medicinal Chemistry, Institute of Pharmacy, Friedrich-Schiller-University Jena, University Hospital Jena, Jena, Germany
| | - Friederike Dehm
- Department of Pharmaceutical/Medicinal Chemistry, Institute of Pharmacy, Friedrich-Schiller-University Jena, University Hospital Jena, Jena, Germany.,Department of Pharmacy, School of Medicine, University of Naples Federico II, Naples, Italy
| | - Antonietta Rossi
- Department of Pharmacy, School of Medicine, University of Naples Federico II, Naples, Italy
| | - Jana Gerstmeier
- Department of Pharmaceutical/Medicinal Chemistry, Institute of Pharmacy, Friedrich-Schiller-University Jena, University Hospital Jena, Jena, Germany
| | - Fabiana Troisi
- Department of Pharmaceutical/Medicinal Chemistry, Institute of Pharmacy, Friedrich-Schiller-University Jena, University Hospital Jena, Jena, Germany
| | - Helmut Pein
- Department of Pharmaceutical/Medicinal Chemistry, Institute of Pharmacy, Friedrich-Schiller-University Jena, University Hospital Jena, Jena, Germany
| | - Anja M Schaible
- Department of Pharmaceutical/Medicinal Chemistry, Institute of Pharmacy, Friedrich-Schiller-University Jena, University Hospital Jena, Jena, Germany
| | - Christina Weinigel
- Institute of Transfusion Medicine, University Hospital Jena, Jena, Germany
| | - Silke Rummler
- Institute of Transfusion Medicine, University Hospital Jena, Jena, Germany
| | - Hinnak Northoff
- Institute for Clinical and Experimental Transfusion Medicine, University Medical Center Tuebingen, and
| | - Stefan Laufer
- Department of Medicinal Chemistry, Pharmaceutical Institute, University Tuebingen, Tuebingen, Germany
| | - Thorsten J Maier
- Aarhus University, Department of Biomedicine and Center for Study and Prevention of Neurodegenerative Inflammation (NEURODIN), Aarhus, Denmark.,Department of Anesthesia, Intensive Care Medicine and Pain Therapy, University Hospital Frankfurt, Frankfurt am Main, Germany
| | - Olof Rådmark
- Department of Medical Biochemistry and Biophysics, Division of Physiological Chemistry II, Karolinska Institutet, Stockholm, Sweden
| | - Bengt Samuelsson
- Department of Medical Biochemistry and Biophysics, Division of Physiological Chemistry II, Karolinska Institutet, Stockholm, Sweden
| | - Andreas Koeberle
- Department of Pharmaceutical/Medicinal Chemistry, Institute of Pharmacy, Friedrich-Schiller-University Jena, University Hospital Jena, Jena, Germany
| | - Lidia Sautebin
- Department of Pharmacy, School of Medicine, University of Naples Federico II, Naples, Italy
| | - Oliver Werz
- Department of Pharmaceutical/Medicinal Chemistry, Institute of Pharmacy, Friedrich-Schiller-University Jena, University Hospital Jena, Jena, Germany
| |
Collapse
|
81
|
Genome-Wide Linkage Analysis of Large Multiple Multigenerational Families Identifies Novel Genetic Loci for Coronary Artery Disease. Sci Rep 2017; 7:5472. [PMID: 28710368 PMCID: PMC5511258 DOI: 10.1038/s41598-017-05381-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2016] [Accepted: 05/30/2017] [Indexed: 01/10/2023] Open
Abstract
Coronary artery disease (CAD) is the leading cause of death, and genetic factors contribute significantly to risk of CAD. This study aims to identify new CAD genetic loci through a large-scale linkage analysis of 24 large and multigenerational families with 433 family members (GeneQuest II). All family members were genotyped with markers spaced by every 10 cM and a model-free nonparametric linkage (NPL-all) analysis was carried out. Two highly significant CAD loci were identified on chromosome 17q21.2 (NPL score of 6.20) and 7p22.2 (NPL score of 5.19). We also identified four loci with significant NPL scores between 4.09 and 4.99 on 2q33.3, 3q29, 5q13.2 and 9q22.33. Similar analyses in individual families confirmed the six significant CAD loci and identified seven new highly significant linkages on 9p24.2, 9q34.2, 12q13.13, 15q26.1, 17q22, 20p12.3, and 22q12.1, and two significant loci on 2q11.2 and 11q14.1. Two loci on 3q29 and 9q22.33 were also successfully replicated in our previous linkage analysis of 428 nuclear families. Moreover, two published risk variants, SNP rs46522 in UBE2Z and SNP rs6725887 in WDR12 by GWAS, were found within the 17q21.2 and 2q33.3 loci. These studies lay a foundation for future identification of causative variants and genes for CAD.
Collapse
|
82
|
Drug discovery approaches targeting 5-lipoxygenase-activating protein (FLAP) for inhibition of cellular leukotriene biosynthesis. Eur J Med Chem 2017; 153:34-48. [PMID: 28784429 DOI: 10.1016/j.ejmech.2017.07.019] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2017] [Revised: 06/29/2017] [Accepted: 07/12/2017] [Indexed: 11/23/2022]
Abstract
Leukotrienes are proinflammatory lipid mediators associated with diverse chronic inflammatory diseases such as asthma, COPD, IBD, arthritis, atherosclerosis, dermatitis and cancer. Cellular leukotrienes are produced from arachidonic acid via the 5-lipoxygenase pathway in which the 5-lipoxygenase activating protein, also named as FLAP, plays a critical role by operating as a regulatory protein for efficient transfer of arachidonic acid to 5-lipoxygenase. By blocking leukotriene production, FLAP inhibitors may behave as broad-spectrum leukotriene modulators, which might be of therapeutic use for chronic inflammatory diseases requiring anti-leukotriene therapy. The early development of FLAP inhibitors (i.e. MK-886, MK-591, BAY-X-1005) mostly concentrated on asthma cure, and resulted in promising readouts in preclinical and clinical studies with asthma patients. Following the recent elucidation of the 3D-structure of FLAP, development of new inhibitor chemotypes is highly accelerated, eventually leading to the evolution of many un-drug-like structures into more drug-like entities such as AZD6642 and BI665915 as development candidates. The most clinically advanced FLAP inhibitor to date is GSK2190918 (formerly AM803) that has successfully completed phase II clinical trials in asthmatics. Concluding, although there are no FLAP inhibitors reached to the drug approval phase yet, due to the rising number of indications for anti-LT therapy such as atherosclerosis, FLAP inhibitor development remains a significant research field. FLAP inhibitors reviewed herein are classified into four sub-classes as the first-generation FLAP inhibitors (indole and quinoline derivatives), the second-generation FLAP inhibitors (diaryl-alkanes and biaryl amino-heteroarenes), the benzimidazole-containing FLAP inhibitors and other FLAP inhibitors with polypharmacology for easiness of the reader. Hence, we meticulously summarize how FLAP inhibitors historically developed from scratch to their current advanced state, and leave the reader with a positive view that a FLAP inhibitor might soon reach to the need of patients who may require anti-LT therapy.
Collapse
|
83
|
Elosua R, Sayols-Baixeras S. The Genetics of Ischemic Heart Disease: From Current Knowledge to Clinical Implications. ACTA ACUST UNITED AC 2017. [PMID: 28623161 DOI: 10.1016/j.rec.2017.02.046] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Ischemic heart disease continues to cause high morbidity and mortality. Its prevalence is expected to increase due to population aging, and its prevention is a major goal of health policies. The risk of developing ischemic heart disease is related to a complex interplay between genetic, environmental, and lifestyle factors. In the last decade, considerable progress has been made in knowledge of the genetic architecture of this disease. This narrative review provides an overview of current knowledge of the genetics of ischemic heart disease and of its translation to clinical practice: identification of new therapeutic targets, assessment of the causal relationship between biomarkers and disease, improved risk prediction, and identification of responders and nonresponders to specific drugs (pharmacogenomics).
Collapse
Affiliation(s)
- Roberto Elosua
- Grupo de Epidemiología y Genética Cardiovascular, Instituto Hospital del Mar de Investigaciones Médicas (IMIM), Barcelona, Spain; Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Barcelona, Spain.
| | - Sergi Sayols-Baixeras
- Grupo de Epidemiología y Genética Cardiovascular, Instituto Hospital del Mar de Investigaciones Médicas (IMIM), Barcelona, Spain; Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Barcelona, Spain; Departamento de Ciencias de la Salud y de la Vida, Universidad Pompeu Fabra, Barcelona, Spain
| |
Collapse
|
84
|
Cysteinyl Leukotrienes as Potential Pharmacological Targets for Cerebral Diseases. Mediators Inflamm 2017; 2017:3454212. [PMID: 28607533 PMCID: PMC5451784 DOI: 10.1155/2017/3454212] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2017] [Revised: 04/10/2017] [Accepted: 04/19/2017] [Indexed: 02/06/2023] Open
Abstract
Cysteinyl leukotrienes (CysLTs) are potent lipid mediators widely known for their actions in asthma and in allergic rhinitis. Accumulating data highlights their involvement in a broader range of inflammation-associated diseases such as cancer, atopic dermatitis, rheumatoid arthritis, and cardiovascular diseases. The reported elevated levels of CysLTs in acute and chronic brain lesions, the association between the genetic polymorphisms in the LTs biosynthesis pathways and the risk of cerebral pathological events, and the evidence from animal models link also CysLTs and brain diseases. This review will give an overview of how far research has gone into the evaluation of the role of CysLTs in the most prevalent neurodegenerative disorders (ischemia, Alzheimer's and Parkinson's diseases, multiple sclerosis/experimental autoimmune encephalomyelitis, and epilepsy) in order to understand the underlying mechanism by which they might be central in the disease progression.
Collapse
|
85
|
Hoxha M, Rovati GE, Cavanillas AB. The leukotriene receptor antagonist montelukast and its possible role in the cardiovascular field. Eur J Clin Pharmacol 2017; 73:799-809. [PMID: 28374082 DOI: 10.1007/s00228-017-2242-2] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2017] [Accepted: 03/22/2017] [Indexed: 12/31/2022]
Abstract
BACKGROUND Cysteinyl leukotrienes (LTC4, LTD4, and LTE4) are pro-inflammatory mediators of the 5-lipooxygenase (5-LO) pathway, that play an important role in bronchoconstriction, but can also enhance endothelial cell permeability and myocardial contractility, and are involved in many other inflammatory conditions. In the late 1990s, leukotriene receptor antagonists (LTRAs) were introduced in therapy for asthma and later on, approved for the relief of the symptoms of allergic rhinitis, chronic obstructive pulmonary disease, and urticaria. In addition, it has been shown that LTRAs may have a potential role in preventing atherosclerosis progression. PURPOSE The aims of this short review are to delineate the potential cardiovascular protective role of a LTRA, montelukast, beyond its traditional use, and to foster the design of appropriate clinical trials to test this hypothesis. RESULTS AND CONCLUSIONS What it is known about leukotriene receptor antagonists? •Leukotriene receptor antagonist, such as montelukast and zafirlukast, is used in asthma, COPD, and allergic rhinitis. • Montelukast is the most prescribed CysLT1 antagonist used in asthmatic patients. • Different in vivo animal studies have shown that leukotriene receptor antagonists can prevent the atherosclerosis progression, and have a protective role after cerebral ischemia. What we still need to know? • Today, there is a need for conducting clinical trials to assess the role of montelukast in reducing cardiovascular risk and to further understand the mechanism of action behind this effect.
Collapse
Affiliation(s)
- Malvina Hoxha
- Department of Chemical, Toxicological and Pharmacological Evaluation of Drugs, Catholic University Our Lady of Good Counsel, Rruga. D. Hoxha, Tirana, Albania.
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Via Balzaretti, 9-20133, Milan, Italy.
| | - G Enrico Rovati
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Via Balzaretti, 9-20133, Milan, Italy
| | - Aurora Bueno Cavanillas
- IBS Granada, University of Granada, CIBER of Epidemiology and Public Health (CIBERESP), Granada, Spain
| |
Collapse
|
86
|
Whole-genome sequencing identifies rare genotypes in COMP and CHADL associated with high risk of hip osteoarthritis. Nat Genet 2017; 49:801-805. [DOI: 10.1038/ng.3816] [Citation(s) in RCA: 64] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2016] [Accepted: 02/23/2017] [Indexed: 12/13/2022]
|
87
|
Lu W, Yao X, Ouyang P, Dong N, Wu D, Jiang X, Wu Z, Zhang C, Xu Z, Tang Y, Zou S, Liu M, Li J, Zeng M, Lin P, Cheng F, Huang J. Drug Repurposing of Histone Deacetylase Inhibitors That Alleviate Neutrophilic Inflammation in Acute Lung Injury and Idiopathic Pulmonary Fibrosis via Inhibiting Leukotriene A4 Hydrolase and Blocking LTB4 Biosynthesis. J Med Chem 2017; 60:1817-1828. [PMID: 28218840 DOI: 10.1021/acs.jmedchem.6b01507] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Acute lung injury (ALI) and idiopathic pulmonary fibrosis (IPF) are both serious public health problems with high incidence and mortality rate in adults, and with few drugs available for the efficient treatment in clinic. In this study, we identified that two known histone deacetylase (HDAC) inhibitors, suberanilohydroxamic acid (SAHA, 1) and its analogue 4-(dimethylamino)-N-[7-(hydroxyamino)-7-oxoheptyl]benzamide (2), are effective inhibitors of Leukotriene A4 hydrolase (LTA4H), a key enzyme in the biosynthesis of leukotriene B4 (LTB4), across a panel of 18 HDAC inhibitors, using enzymatic assay, thermofluor assay, and X-ray crystallographic investigation. Importantly, both 1 and 2 markedly diminish early neutrophilic inflammation in mouse models of ALI and IPF under a clinical safety dose. Detailed mechanisms of down-regulation of proinflammatory cytokines by 1 or 2 were determined in vivo. Collectively, 1 and 2 would provide promising agents with well-known clinical safety for potential treatment in patients with ALI and IPF via pharmacologically inhibiting LAT4H and blocking LTB4 biosynthesis.
Collapse
Affiliation(s)
- Weiqiang Lu
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology , Shanghai 200237, China.,Shanghai Key Laboratory of Regulatory Biology, The Institute of Biomedical Sciences and School of Life Sciences, East China Normal University , Shanghai 200241, China
| | - Xue Yao
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology , Shanghai 200237, China
| | - Ping Ouyang
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology , Shanghai 200237, China
| | - Ningning Dong
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology , Shanghai 200237, China
| | - Dang Wu
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology , Shanghai 200237, China
| | - Xingwu Jiang
- Shanghai Key Laboratory of Regulatory Biology, The Institute of Biomedical Sciences and School of Life Sciences, East China Normal University , Shanghai 200241, China
| | - Zengrui Wu
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology , Shanghai 200237, China
| | - Chen Zhang
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology , Shanghai 200237, China
| | - Zhongyu Xu
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology , Shanghai 200237, China
| | - Yun Tang
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology , Shanghai 200237, China
| | - Shien Zou
- Department of Gynecology, Obstetrics and Gynecology Hospital of Fudan University , Shanghai 200011, China
| | - Mingyao Liu
- Shanghai Key Laboratory of Regulatory Biology, The Institute of Biomedical Sciences and School of Life Sciences, East China Normal University , Shanghai 200241, China
| | - Jian Li
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology , Shanghai 200237, China
| | - Minghua Zeng
- Key Laboratory for the Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education), School of Chemistry & Chemical Engineering, Guangxi Normal University , Guilin 541004, China
| | - Ping Lin
- State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy , Chengdu 610041, Sichuan, China
| | - Feixiong Cheng
- State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy , Chengdu 610041, Sichuan, China.,Center for Complex Networks Research, Northeastern University , Boston, Massachusetts 02115, United States.,Center for Cancer Systems Biology and Department of Cancer Biology, Dana-Farber Cancer Institute, Harvard Medical School , Boston, Massachusetts 02215, United States
| | - Jin Huang
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology , Shanghai 200237, China
| |
Collapse
|
88
|
Discovery of the first dual inhibitor of the 5-lipoxygenase-activating protein and soluble epoxide hydrolase using pharmacophore-based virtual screening. Sci Rep 2017; 7:42751. [PMID: 28218273 PMCID: PMC5317001 DOI: 10.1038/srep42751] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2016] [Accepted: 01/13/2017] [Indexed: 11/09/2022] Open
Abstract
Leukotrienes (LTs) are pro-inflammatory lipid mediators derived from arachidonic acid (AA) with roles in inflammatory and allergic diseases. The biosynthesis of LTs is initiated by transfer of AA via the 5-lipoxygenase-activating protein (FLAP) to 5-lipoxygenase (5-LO). FLAP inhibition abolishes LT formation exerting anti-inflammatory effects. The soluble epoxide hydrolase (sEH) converts AA-derived anti-inflammatory epoxyeicosatrienoic acids (EETs) to dihydroxyeicosatetraenoic acids (di-HETEs). Its inhibition consequently also counteracts inflammation. Targeting both LT biosynthesis and the conversion of EETs with a dual inhibitor of FLAP and sEH may represent a novel, powerful anti-inflammatory strategy. We present a pharmacophore-based virtual screening campaign that led to 20 hit compounds of which 4 targeted FLAP and 4 were sEH inhibitors. Among them, the first dual inhibitor for sEH and FLAP was identified, N-[4-(benzothiazol-2-ylmethoxy)-2-methylphenyl]-N'-(3,4-dichlorophenyl)urea with IC50 values of 200 nM in a cell-based FLAP test system and 20 nM for sEH activity in a cell-free assay.
Collapse
|
89
|
Kowal-Bielecka O, Chwiesko-Minarowska S, Bernatowicz PL, Allanore Y, Radstake T, Matucci-Cerinic M, Broen J, Hesselstrand R, Krasowska D, Riemekasten G, Vonk M, Kowalczuk O, Bielecki M, Milewski R, Chyczewski L, Niklinski J, Kowal K. The arachidonate 5-lipoxygenase activating protein gene polymorphism is associated with the risk of scleroderma-related interstitial lung disease: a multicentre European Scleroderma Trials and Research group (EUSTAR) study. Rheumatology (Oxford) 2017; 56:844-852. [DOI: 10.1093/rheumatology/kew499] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2016] [Indexed: 01/02/2023] Open
|
90
|
Chen Z, Zheng J, Liu W, Yang K, Li K, Huang B, Zhu R, Lu X, Li L. The SG13S114 polymorphism of the ALOX5AP gene is associated with ischemic stroke in Europeans: a meta-analysis of 8062 subjects. Neurol Sci 2017; 38:579-587. [PMID: 28101761 DOI: 10.1007/s10072-016-2804-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2016] [Accepted: 12/22/2016] [Indexed: 10/20/2022]
Abstract
The association between ALOX5AP SG13S114 polymorphism and ischemic stroke (IS) susceptibility has extensively been investigated, especially in white populations; however, the results were inconclusive. Here, we perform a meta-analysis to clarify the effect of SG13S114 variant on the IS risk in Europeans. The Web of Science, PubMed, EMBASE, and Medline were searched up to August 1st, 2016. Data were extracted and the odd ratios (ORs) and 95% confidence intervals (CIs) were calculated by a fixed-effects or random-effects model. In total, 8 case control studies involved 8062 subjects were finally included in this meta-analysis. We observed a significantly decreased IS risk in persons carrying an A allele at the SG13S114 polymorphism compared with those with a T allele (A vs T, OR = 0.856, 95% CI = 0.797-0.919, p < 0.001). In addition, the results of sensitivity and cumulative meta-analysis indicated the robustness of our results. In addition, the publication bias was not detected using the funnel plot and Egger's tests. In summary, the present meta-analysis suggested that the A allele at the ALOX5AP SG13S114 polymorphism is a protective factor for the IS in the Europeans. In addition, further studies with large sample size are needed to validate the association, as well as in other ethnic groups.
Collapse
Affiliation(s)
- Zhongjun Chen
- Department of Neurosurgery, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029, Jiangsu, China.,Department of Neurosurgery, The Affiliated Huai'an Hospital of Xuzhou Medical College, No. 62 South Huaihai Road, Huai'an, 223002, China
| | - Jinyu Zheng
- Department of Neurosurgery, The Affiliated Huai'an Hospital of Xuzhou Medical College, No. 62 South Huaihai Road, Huai'an, 223002, China
| | - Wenguang Liu
- Department of Neurosurgery, The Affiliated Huai'an Hospital of Xuzhou Medical College, No. 62 South Huaihai Road, Huai'an, 223002, China
| | - Kun Yang
- Department of Neurosurgery, Nanjing Brain Hospital Affiliated with Nanjing Medical University, 264 Guangzhou Road, Nanjing, 210029, Jiangsu, China
| | - Kai Li
- Department of Neurosurgery, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, 210011, Jiangsu, China
| | - Baosheng Huang
- Department of Neurosurgery, Sir Shaw Hospital, Nanjing Medical University, Nanjing, 211100, Jiangsu, China
| | - Ronglan Zhu
- Department of Neurosurgery, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029, Jiangsu, China
| | - Xiaocheng Lu
- Department of Neurosurgery, The First Affiliated Hospital of Soochow University, Suzhou, 215006, China
| | - Lixin Li
- Department of Neurosurgery, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029, Jiangsu, China.
| |
Collapse
|
91
|
Common Polymorphisms in the 5-Lipoxygenase Pathway and Risk of Incident Myocardial Infarction: A Danish Case-Cohort Study. PLoS One 2016; 11:e0167217. [PMID: 27893808 PMCID: PMC5125697 DOI: 10.1371/journal.pone.0167217] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2016] [Accepted: 11/10/2016] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND The 5-lipoxygenase pathway (5-LOX) has been implicated in the development of cardiovascular disease and studies have suggested that genetic polymorphisms related to key enzymes in this pathway may confer risk of myocardial infarction (MI). This study investigated the association of pre-selected genetic polymorphisms in four candidate genes of 5-LOX (arachidonate 5-lipoxygenase and its activating protein (ALOX-5 and FLAP), leukotriene A4 hydroxylase (LTA4-H) and leukotriene C4 synthase (LTC4-S)) with incident MI. METHODS In a Danish cohort including 57,053 participants, aged 50-64 at enrolment and recruited from 1993-97, we conducted a case-cohort study including cases with incident MI and a randomly selected sub cohort of 3,000 participants. Cases were identified from national registries through July 2013. A total of 22 SNPs were selected and genotyped using the commercially available KASP™ assay. A tandem-repeat polymorphism, located in the ALOX-5 gene, was genotyped by multi-titre plate sequencing. Haplotypes were inferred using PHASE 2.1. RESULTS During a median follow-up of 17.0 years we identified 3,089 cases of incident MI. In FLAP, two SNPs were negatively associated with incident MI (rs9551963 & rs17222842) while one SNP (rs2247570) located in LTA4-H, was associated with higher risk of MI when comparing subjects with two copies of the variant allele to homozygotes for the wild type. However, only rs17222842 remained significantly associated with MI after correcting for multiple testing. Furthermore, the promoter polymorphism rs59439148 was associated with risk of MI in men. For male carriers of two variant alleles we found a hazard ratio of 1.63 (95% CI: 1.06;2.52) compared to homozygotes for the wild type. Previously described haplotypes (Hap-A -B, -E and -K) were not associated with MI in our population. CONCLUSION In conclusion, some common polymorphisms in the 5-lipoxygenase pathway were modestly associated with incident MI, suggesting a potential role for this pathway in the development of cardiovascular disease.
Collapse
|
92
|
Abstract
INTRODUCTION Preeclampsia is a major pregnancy disease, explained partly by genetic predispositions. STOX1, a transcription factor discovered in 2005, was the first gene directly associated with genetic forms of the disease. Alterations of STOX1 expression as well as STOX1 variants have also been associated to Alzheimer's disease. These observations make of this gene a putative therapeutic target. Area covered: Two major isoforms (STOX1A and STOX1B) are encoded by the gene and are theoretically able to compete for the same binding site, while only the most complete (STOX1A) is supposed to be able to activate gene expression. This makes the ratio between STOX1A and STOX1B as well as their position inside the cell (nucleus or cytoplasm) crucial to understand how STOX1 functions. STOX1 appears to have multiple gene targets, especially in pathways connected to inflammation, oxidative stress, and cell cycle. Expert opinion: STOX1-directed therapies, could be directed either towards its targets (genes or pathways), or directly at STOX1. For this the addressing of STOX1 to various cell compartments could theoretically be modified; also it could be possible of altering the balance between the two isoforms, through selectively inhibiting one of them, possibly improving the outcomes in severe preeclampsia.
Collapse
Affiliation(s)
- Daniel Vaiman
- a Department of Development, Reproduction and Cancer , Institut Cochin , Paris , France
| | - Francisco Miralles
- a Department of Development, Reproduction and Cancer , Institut Cochin , Paris , France
| |
Collapse
|
93
|
Iribarren C, Lu M, Jorgenson E, Martínez M, Lluis-Ganella C, Subirana I, Salas E, Elosua R. Clinical Utility of Multimarker Genetic Risk Scores for Prediction of Incident Coronary Heart Disease: A Cohort Study Among Over 51 000 Individuals of European Ancestry. ACTA ACUST UNITED AC 2016; 9:531-540. [PMID: 27780846 DOI: 10.1161/circgenetics.116.001522] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2016] [Accepted: 09/28/2016] [Indexed: 12/28/2022]
Abstract
BACKGROUND We evaluated whether including multilocus genetic risk scores (GRSs) into the Framingham Risk Equation improves the predictive capacity, discrimination, and reclassification of asymptomatic individuals with respect to coronary heart disease (CHD) risk. METHODS AND RESULTS We performed a cohort study among 51 954 European-ancestry members of a Northern California integrated healthcare system (67% female; mean age 59) free of CHD at baseline (2007-2008). Four GRSs were constructed using between 8 and 51 previously identified genetic variants. After a mean (±SD) follow-up of 5.9 (±1.5) years, 1864 incident CHD events were documented. All GRSs were linearly associated with CHD in a model adjusted by individual risk factors: hazard ratio (95% confidence interval) per SD unit: 1.21 (1.15-1.26) for GRS_8, 1.20 (1.15-1.26) for GRS_12, 1.23 (1.17-1.28) for GRS_36, and 1.23 (1.17-1.28) for GRS_51. Inclusion of the GRSs improved the C statistic (ΔC statistic =0.008 for GRS_8 and GRS_36; 0.007 for GRS_12; and 0.009 for GRS_51; all P<0.001). The net reclassification improvement was 5% for GRS_8, GRS_12, and GRS_36 and 4% for GRS_51 in the entire cohort and was (after correcting for bias) 9% for GRS_8 and GRS_12 and 7% for GRS_36 and GRS_51 when analyzing those classified as intermediate Framingham risk (10%-20%). The number required to treat to prevent 1 CHD after selectively treating with statins up-reclassified subjects on the basis of genetic information was 36 for GRS_8 and GRS_12, 41 for GRS_36, and 43 for GRS_51. CONCLUSIONS Our results demonstrate significant and clinically relevant incremental discriminative/predictive capability of 4 multilocus GRSs for incident CHD among subjects of European ancestry.
Collapse
Affiliation(s)
- Carlos Iribarren
- From the Kaiser Permanente Northern California Division of Research, Oakland, CA (C.I., M.L., E.J.); Gendiag, Inc/Ferrer inCode, Inc, Barcelona, Spain (M.M., C.L.-G., E.S.); CIBER of Epidemiology and Public Health, Barcelona, Spain (I.S.); and Cardiovascular Epidemiology & Genetics, IMIM, Barcelona, Spain (I.S., R.E.).
| | - Meng Lu
- From the Kaiser Permanente Northern California Division of Research, Oakland, CA (C.I., M.L., E.J.); Gendiag, Inc/Ferrer inCode, Inc, Barcelona, Spain (M.M., C.L.-G., E.S.); CIBER of Epidemiology and Public Health, Barcelona, Spain (I.S.); and Cardiovascular Epidemiology & Genetics, IMIM, Barcelona, Spain (I.S., R.E.)
| | - Eric Jorgenson
- From the Kaiser Permanente Northern California Division of Research, Oakland, CA (C.I., M.L., E.J.); Gendiag, Inc/Ferrer inCode, Inc, Barcelona, Spain (M.M., C.L.-G., E.S.); CIBER of Epidemiology and Public Health, Barcelona, Spain (I.S.); and Cardiovascular Epidemiology & Genetics, IMIM, Barcelona, Spain (I.S., R.E.)
| | - Manuel Martínez
- From the Kaiser Permanente Northern California Division of Research, Oakland, CA (C.I., M.L., E.J.); Gendiag, Inc/Ferrer inCode, Inc, Barcelona, Spain (M.M., C.L.-G., E.S.); CIBER of Epidemiology and Public Health, Barcelona, Spain (I.S.); and Cardiovascular Epidemiology & Genetics, IMIM, Barcelona, Spain (I.S., R.E.)
| | - Carla Lluis-Ganella
- From the Kaiser Permanente Northern California Division of Research, Oakland, CA (C.I., M.L., E.J.); Gendiag, Inc/Ferrer inCode, Inc, Barcelona, Spain (M.M., C.L.-G., E.S.); CIBER of Epidemiology and Public Health, Barcelona, Spain (I.S.); and Cardiovascular Epidemiology & Genetics, IMIM, Barcelona, Spain (I.S., R.E.)
| | - Isaac Subirana
- From the Kaiser Permanente Northern California Division of Research, Oakland, CA (C.I., M.L., E.J.); Gendiag, Inc/Ferrer inCode, Inc, Barcelona, Spain (M.M., C.L.-G., E.S.); CIBER of Epidemiology and Public Health, Barcelona, Spain (I.S.); and Cardiovascular Epidemiology & Genetics, IMIM, Barcelona, Spain (I.S., R.E.)
| | - Eduardo Salas
- From the Kaiser Permanente Northern California Division of Research, Oakland, CA (C.I., M.L., E.J.); Gendiag, Inc/Ferrer inCode, Inc, Barcelona, Spain (M.M., C.L.-G., E.S.); CIBER of Epidemiology and Public Health, Barcelona, Spain (I.S.); and Cardiovascular Epidemiology & Genetics, IMIM, Barcelona, Spain (I.S., R.E.)
| | - Roberto Elosua
- From the Kaiser Permanente Northern California Division of Research, Oakland, CA (C.I., M.L., E.J.); Gendiag, Inc/Ferrer inCode, Inc, Barcelona, Spain (M.M., C.L.-G., E.S.); CIBER of Epidemiology and Public Health, Barcelona, Spain (I.S.); and Cardiovascular Epidemiology & Genetics, IMIM, Barcelona, Spain (I.S., R.E.)
| |
Collapse
|
94
|
Lang L, Dong N, Wu D, Yao X, Lu W, Zhang C, Ouyang P, Zhu J, Tang Y, Wang W, Li J, Huang J. 2-Arylbenzo[b]furan derivatives as potent human lipoxygenase inhibitors. J Enzyme Inhib Med Chem 2016; 31:98-105. [DOI: 10.1080/14756366.2016.1220376] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Affiliation(s)
- Li Lang
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai, China,
| | - Ningning Dong
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai, China,
| | - Deyan Wu
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai, China,
| | - Xue Yao
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai, China,
| | - Weiqiang Lu
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, China, and
| | - Chen Zhang
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai, China,
| | - Ping Ouyang
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai, China,
| | - Jin Zhu
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai, China,
| | - Yun Tang
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai, China,
| | - Wei Wang
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai, China,
- Department of Chemistry & Chemical Biology, University of New Mexico, Albuquerque, NM, USA
| | - Jian Li
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai, China,
| | - Jin Huang
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai, China,
| |
Collapse
|
95
|
Garscha U, Voelker S, Pace S, Gerstmeier J, Emini B, Liening S, Rossi A, Weinigel C, Rummler S, Schubert US, Scriba GKE, Çelikoğlu E, Çalışkan B, Banoglu E, Sautebin L, Werz O. BRP-187: A potent inhibitor of leukotriene biosynthesis that acts through impeding the dynamic 5-lipoxygenase/5-lipoxygenase-activating protein (FLAP) complex assembly. Biochem Pharmacol 2016; 119:17-26. [PMID: 27592027 DOI: 10.1016/j.bcp.2016.08.023] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2016] [Accepted: 08/29/2016] [Indexed: 11/19/2022]
Abstract
The pro-inflammatory leukotrienes (LTs) are formed from arachidonic acid (AA) in activated leukocytes, where 5-lipoxygenase (5-LO) translocates to the nuclear envelope to assemble a functional complex with the integral nuclear membrane protein 5-LO-activating protein (FLAP). FLAP, a MAPEG family member, facilitates AA transfer to 5-LO for efficient conversion, and LT biosynthesis critically depends on FLAP. Here we show that the novel LT biosynthesis inhibitor BRP-187 prevents the 5-LO/FLAP interaction at the nuclear envelope of human leukocytes without blocking 5-LO nuclear redistribution. BRP-187 inhibited 5-LO product formation in human monocytes and polymorphonuclear leukocytes stimulated by lipopolysaccharide plus N-formyl-methionyl-leucyl-phenylalanine (IC50=7-10nM), and upon activation by ionophore A23187 (IC50=10-60nM). Excess of exogenous AA markedly impaired the potency of BRP-187. Direct 5-LO inhibition in cell-free assays was evident only at >35-fold higher concentrations, which was reversible and not improved under reducing conditions. BRP-187 prevented A23187-induced 5-LO/FLAP complex assembly in leukocytes but failed to block 5-LO nuclear translocation, features that were shared with the FLAP inhibitor MK886. Whereas AA release, cyclooxygenases and related LOs were unaffected, BRP-187 also potently inhibited microsomal prostaglandin E2 synthase-1 (IC50=0.2μM), another MAPEG member. In vivo, BRP-187 (10mg/kg) exhibited significant effectiveness in zymosan-induced murine peritonitis, suppressing LT levels in peritoneal exudates as well as vascular permeability and neutrophil infiltration. Together, BRP-187 potently inhibits LT biosynthesis in vitro and in vivo, which seemingly is caused by preventing the 5-LO/FLAP complex assembly and warrants further preclinical evaluation.
Collapse
Affiliation(s)
- Ulrike Garscha
- Chair of Pharmaceutical/Medicinal Chemistry, Institute of Pharmacy, Friedrich Schiller University Jena, Philosophenweg 14, D-07743 Jena, Germany.
| | - Susanna Voelker
- Chair of Pharmaceutical/Medicinal Chemistry, Institute of Pharmacy, Friedrich Schiller University Jena, Philosophenweg 14, D-07743 Jena, Germany.
| | - Simona Pace
- Chair of Pharmaceutical/Medicinal Chemistry, Institute of Pharmacy, Friedrich Schiller University Jena, Philosophenweg 14, D-07743 Jena, Germany.
| | - Jana Gerstmeier
- Chair of Pharmaceutical/Medicinal Chemistry, Institute of Pharmacy, Friedrich Schiller University Jena, Philosophenweg 14, D-07743 Jena, Germany.
| | - Besa Emini
- Chair of Pharmaceutical/Medicinal Chemistry, Institute of Pharmacy, Friedrich Schiller University Jena, Philosophenweg 14, D-07743 Jena, Germany.
| | - Stefanie Liening
- Chair of Pharmaceutical/Medicinal Chemistry, Institute of Pharmacy, Friedrich Schiller University Jena, Philosophenweg 14, D-07743 Jena, Germany.
| | - Antonietta Rossi
- Department of Pharmacy, University of Naples Federico II, 80131 Naples, Italy.
| | - Christina Weinigel
- Institute of Transfusion Medicine, University Hospital Jena, 07743 Jena, Germany.
| | - Silke Rummler
- Institute of Transfusion Medicine, University Hospital Jena, 07743 Jena, Germany.
| | - Ulrich S Schubert
- Jena Center for Soft Matter (JCSM), Friedrich Schiller University Jena, Philosophenweg 7, 07743 Jena, Germany; Laboratory of Organic and Macromolecular Chemistry (IOMC), Friedrich Schiller University Jena, Humboldtstrasse 10, D-07743 Jena, Germany.
| | - Gerhard K E Scriba
- Chair of Pharmaceutical/Medicinal Chemistry, Institute of Pharmacy, Friedrich Schiller University Jena, Philosophenweg 14, D-07743 Jena, Germany.
| | - Erşan Çelikoğlu
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Gazi University, Etiler, 06330 Yenimahalle, Ankara, Turkey.
| | - Burcu Çalışkan
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Gazi University, Etiler, 06330 Yenimahalle, Ankara, Turkey.
| | - Erden Banoglu
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Gazi University, Etiler, 06330 Yenimahalle, Ankara, Turkey.
| | - Lidia Sautebin
- Department of Pharmacy, University of Naples Federico II, 80131 Naples, Italy.
| | - Oliver Werz
- Chair of Pharmaceutical/Medicinal Chemistry, Institute of Pharmacy, Friedrich Schiller University Jena, Philosophenweg 14, D-07743 Jena, Germany; Jena Center for Soft Matter (JCSM), Friedrich Schiller University Jena, Philosophenweg 7, 07743 Jena, Germany.
| |
Collapse
|
96
|
Yang D, Huang X, Cui C, Zhang Y, Li Y, Zang X, He Y, Zheng H. Genetic Variants in the Transcriptional Regulatory Region of the ALOX5AP gene and Susceptibility to Ischemic Stroke in Chinese Populations. Sci Rep 2016; 6:29513. [PMID: 27416969 PMCID: PMC4945871 DOI: 10.1038/srep29513] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2015] [Accepted: 06/17/2016] [Indexed: 11/09/2022] Open
Abstract
No coding sequence variants of the ALOX5AP gene that lead to amino acid substitutions have been identified. A two-stage study design was used to explore the relationship between variants in the transcriptional regulatory region of ALOX5AP gene and ischemic stroke (IS) risk in Chinese populations. IS was determined using CT and/or MRI. First, 18 SNPs, located in the upstream promoter region of ALOX5AP gene, were genotyped in 200 IS patients and 200 controls. And one potential associated SNP (rs17222919) was identified (P = 0.005,OR = 0.623, 95% CI: 0.448~0.866). Next, another independent case-control cohort comprising 810 IS patients and 825 matched controls was recruited to investigate the role of rs17222919, rs9579646 polymorphisms and their haplotypes in IS risk. The G allele frequency of rs17222919 in the IS group was significantly lower than that in control group (P = 0.007, OR = 0.792, 95% CI: 0.669~0.937). T-A and G-A haplotypes were associated with IS (P = 0.001,OR = 1.282, 95% CI:1.100~1.495; P = 0.0001, OR = 0.712, 95% CI: 0.598~0.848; respectively). Our study providesevidence that rs17222919 is a potential genetic protective factor against IS. Furthermore, the T-A haplotype is a risk factor and the G-A haplotype is a protective factor against IS in Chinese population.
Collapse
Affiliation(s)
- Dongzhi Yang
- School of life sciences of Zhengzhou University, Zhengzhou, 450052, China
| | - Xiangnan Huang
- School of life sciences of Zhengzhou University, Zhengzhou, 450052, China
| | - Chuanju Cui
- Department of Neurology, The First People's Hospital of Zhengzhou, Zhengzhou, 450000, China
| | - Yuchao Zhang
- Department of Medical Genetics &Cell Biology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450052, China
| | - Ya Li
- Department of Medical Genetics &Cell Biology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450052, China
| | - Xin Zang
- School of life sciences of Zhengzhou University, Zhengzhou, 450052, China
| | - Ying He
- Department of Medical Genetics &Cell Biology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450052, China
| | - Hong Zheng
- Department of Medical Genetics &Cell Biology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450052, China
| |
Collapse
|
97
|
Maga P, Sanak M, Rewerska B, Maga M, Jawien J, Wachsmann A, Rewerski P, Szczeklik W, Celejewska-Wójcik N. Urinary cysteinyl leukotrienes in one-year follow-up of percutaneous transluminal angioplasty for peripheral arterial occlusive disease. Atherosclerosis 2016; 249:174-80. [DOI: 10.1016/j.atherosclerosis.2016.04.013] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/29/2015] [Revised: 03/17/2016] [Accepted: 04/13/2016] [Indexed: 01/04/2023]
|
98
|
Sugimoto MA, Sousa LP, Pinho V, Perretti M, Teixeira MM. Resolution of Inflammation: What Controls Its Onset? Front Immunol 2016. [PMID: 27199985 DOI: 10.3389/fimmu.2016.00.00160] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/04/2023] Open
Abstract
An effective resolution program may be able to prevent the progression from non-resolving acute inflammation to persistent chronic inflammation. It has now become evident that coordinated resolution programs initiate shortly after inflammatory responses begin. In this context, several mechanisms provide the fine-tuning of inflammation and create a favorable environment for the resolution phase to take place and for homeostasis to return. In this review, we focus on the events required for an effective transition from the proinflammatory phase to the onset and establishment of resolution. We suggest that several mediators that promote the inflammatory phase of inflammation can simultaneously initiate a program for active resolution. Indeed, several events enact a decrease in the local chemokine concentration, a reduction which is essential to inhibit further infiltration of neutrophils into the tissue. Interestingly, although neutrophils are cells that characteristically participate in the active phase of inflammation, they also contribute to the onset of resolution. Further understanding of the molecular mechanisms that initiate resolution may be instrumental to develop pro-resolution strategies to treat complex chronic inflammatory diseases, in humans. The efforts to develop strategies based on resolution of inflammation have shaped a new area of pharmacology referred to as "resolution pharmacology."
Collapse
Affiliation(s)
- Michelle A Sugimoto
- Laboratório de Sinalização Inflamação, Departamento de Análises Clínicas e Toxicológicas, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil; Laboratório de Imunofarmacologia, Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Lirlândia P Sousa
- Laboratório de Sinalização Inflamação, Departamento de Análises Clínicas e Toxicológicas, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil; Laboratório de Imunofarmacologia, Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Vanessa Pinho
- Laboratório de Imunofarmacologia, Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil; Laboratório de Resolução da Resposta Inflamatória, Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Mauro Perretti
- William Harvey Research Institute, Barts and The London School of Medicine, Queen Mary University of London , London , UK
| | - Mauro M Teixeira
- Laboratório de Imunofarmacologia, Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais , Belo Horizonte , Brazil
| |
Collapse
|
99
|
DIAKITE BREHIMA, HAMZI KHALIL, HMIMECH WIAM, NADIFI SELLAMA, GMRAVC. Genetic polymorphisms of T-1131C APOA5 and ALOX5AP SG13S114 with the susceptibility of ischaemic stroke in Morocco. J Genet 2016; 95:303-9. [DOI: 10.1007/s12041-016-0635-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
100
|
Sugimoto MA, Sousa LP, Pinho V, Perretti M, Teixeira MM. Resolution of Inflammation: What Controls Its Onset? Front Immunol 2016; 7:160. [PMID: 27199985 PMCID: PMC4845539 DOI: 10.3389/fimmu.2016.00160] [Citation(s) in RCA: 409] [Impact Index Per Article: 51.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2016] [Accepted: 04/12/2016] [Indexed: 12/12/2022] Open
Abstract
An effective resolution program may be able to prevent the progression from non-resolving acute inflammation to persistent chronic inflammation. It has now become evident that coordinated resolution programs initiate shortly after inflammatory responses begin. In this context, several mechanisms provide the fine-tuning of inflammation and create a favorable environment for the resolution phase to take place and for homeostasis to return. In this review, we focus on the events required for an effective transition from the proinflammatory phase to the onset and establishment of resolution. We suggest that several mediators that promote the inflammatory phase of inflammation can simultaneously initiate a program for active resolution. Indeed, several events enact a decrease in the local chemokine concentration, a reduction which is essential to inhibit further infiltration of neutrophils into the tissue. Interestingly, although neutrophils are cells that characteristically participate in the active phase of inflammation, they also contribute to the onset of resolution. Further understanding of the molecular mechanisms that initiate resolution may be instrumental to develop pro-resolution strategies to treat complex chronic inflammatory diseases, in humans. The efforts to develop strategies based on resolution of inflammation have shaped a new area of pharmacology referred to as “resolution pharmacology.”
Collapse
Affiliation(s)
- Michelle A Sugimoto
- Laboratório de Sinalização Inflamação, Departamento de Análises Clínicas e Toxicológicas, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil; Laboratório de Imunofarmacologia, Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Lirlândia P Sousa
- Laboratório de Sinalização Inflamação, Departamento de Análises Clínicas e Toxicológicas, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil; Laboratório de Imunofarmacologia, Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Vanessa Pinho
- Laboratório de Imunofarmacologia, Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil; Laboratório de Resolução da Resposta Inflamatória, Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Mauro Perretti
- William Harvey Research Institute, Barts and The London School of Medicine, Queen Mary University of London , London , UK
| | - Mauro M Teixeira
- Laboratório de Imunofarmacologia, Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais , Belo Horizonte , Brazil
| |
Collapse
|