1
|
Marroquin TY, Guauque-Olarte S. Integrative analysis of gene and protein expression in atherosclerosis-related pathways modulated by periodontal pathogens. Systematic review. JAPANESE DENTAL SCIENCE REVIEW 2023; 59:8-22. [PMID: 36654677 PMCID: PMC9841036 DOI: 10.1016/j.jdsr.2022.12.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Revised: 12/12/2022] [Accepted: 12/15/2022] [Indexed: 01/11/2023] Open
Abstract
The mechanisms modulated by periodontal pathogens in atherosclerosis are not fully understood. Aim: to perform an integrative analysis of gene and protein expression modulated by periodontal pathogens in cells and animal models for atherosclerosis. Methods Cochrane, PRISMA and AMSTAR2 guidelines for systematic reviews were followed. Data search was conducted in Pub-med, LILACS and Science Direct databases. Gene and protein expression data were collected from the included papers to perform an overrepresentation analysis using the Reactome Pathway Analysis tool and the KEGG database. Results Thirty-two papers were included in the review, they analyzed the effect of Fusobacterium nucleatum, Porphyromonas gingivalis, Streptococcus anginosus, Streptococcus sanguinis, Tannerella forsythia, and Treponema denticola or/and their virulent factors on gene and protein expression in human cells and animal models of atherosclerosis. Some of the modulated pathways include the immune system, programmed cell death, cellular responses to external stimuli, transport of small molecules, and signal transduction (p < 0.05). Those pathways are known to be involved in different stages of atherosclerosis progression. Conclusion Based on the performed analysis, it is possible to state that periodontal pathogens have the potential to be a contributing factor for atherosclerosis even in absence of a high-fat diet or high shear stress.
Collapse
Affiliation(s)
| | - Sandra Guauque-Olarte
- GIOM group, Faculty of Dentistry, Universidad Cooperativa de Colombia, Envigado, Colombia
| |
Collapse
|
2
|
Kott KA, Chan AS, Vernon ST, Hansen T, Kim T, de Dreu M, Gunasegaran B, Murphy AJ, Patrick E, Psaltis PJ, Grieve SM, Yang JY, Fazekas de St Groth B, McGuire HM, Figtree GA. Mass cytometry analysis reveals altered immune profiles in patients with coronary artery disease. Clin Transl Immunology 2023; 12:e1462. [PMID: 37927302 PMCID: PMC10621005 DOI: 10.1002/cti2.1462] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Revised: 05/09/2023] [Accepted: 08/09/2023] [Indexed: 11/07/2023] Open
Abstract
Objective The importance of inflammation in atherosclerosis is well accepted, but the role of the adaptive immune system is not yet fully understood. To further explore this, we assessed the circulating immune cell profile of patients with coronary artery disease (CAD) to identify discriminatory features by mass cytometry. Methods Mass cytometry was performed on patient samples from the BioHEART-CT study, gated to detect 82 distinct cell subsets. CT coronary angiograms were analysed to categorise patients as having CAD (CAD+) or having normal coronary arteries (CAD-). Results The discovery cohort included 117 patients (mean age 61 ± 12 years, 49% female); 79 patients (68%) were CAD+. Mass cytometry identified changes in 15 T-cell subsets, with higher numbers of proliferating, highly differentiated and cytotoxic cells and decreases in naïve T cells. Five T-regulatory subsets were related to an age and gender-independent increase in the odds of CAD incidence when expressing CCR2 (OR 1.12), CCR4 (OR 1.08), CD38 and CD45RO (OR 1.13), HLA-DR (OR 1.06) and Ki67 (OR 1.22). Markers of proliferation and differentiation were also increased within B cells, while plasmacytoid dendritic cells were decreased. This combination of changes was assessed using SVM models in discovery and validation cohorts (area under the curve = 0.74 for both), confirming the robust nature of the immune signature detected. Conclusion We identified differences within immune subpopulations of CAD+ patients which are indicative of a systemic immune response to coronary atherosclerosis. This immune signature needs further study via incorporation into risk scoring tools for the precision diagnosis of CAD.
Collapse
Affiliation(s)
- Katharine A Kott
- Cardiothoracic and Vascular HealthKolling Institute of Medical ResearchSydneyNSWAustralia
- Department of Cardiology, Royal North Shore HospitalNorthern Sydney Local Health DistrictSydneyNSWAustralia
- Northern Clinical School, Faculty of Medicine and HealthUniversity of SydneySydneyNSWAustralia
| | - Adam S Chan
- School of Mathematics and StatisticsUniversity of SydneySydneyNSWAustralia
- Charles Perkins CentreUniversity of SydneySydneyNSWAustralia
| | - Stephen T Vernon
- Cardiothoracic and Vascular HealthKolling Institute of Medical ResearchSydneyNSWAustralia
- Department of Cardiology, Royal North Shore HospitalNorthern Sydney Local Health DistrictSydneyNSWAustralia
- Northern Clinical School, Faculty of Medicine and HealthUniversity of SydneySydneyNSWAustralia
| | - Thomas Hansen
- Cardiothoracic and Vascular HealthKolling Institute of Medical ResearchSydneyNSWAustralia
| | - Taiyun Kim
- School of Mathematics and StatisticsUniversity of SydneySydneyNSWAustralia
- Charles Perkins CentreUniversity of SydneySydneyNSWAustralia
| | - Macha de Dreu
- School of Medical Sciences, Faculty of Medicine and HealthUniversity of SydneySydneyNSWAustralia
| | - Bavani Gunasegaran
- Charles Perkins CentreUniversity of SydneySydneyNSWAustralia
- School of Medical Sciences, Faculty of Medicine and HealthUniversity of SydneySydneyNSWAustralia
| | | | - Ellis Patrick
- School of Mathematics and StatisticsUniversity of SydneySydneyNSWAustralia
- Charles Perkins CentreUniversity of SydneySydneyNSWAustralia
| | | | - Stuart M Grieve
- Charles Perkins CentreUniversity of SydneySydneyNSWAustralia
- School of Medical Sciences, Faculty of Medicine and HealthUniversity of SydneySydneyNSWAustralia
- Department of RadiologyRoyal Prince Alfred HospitalSydneyNSWAustralia
- Imaging and Phenotyping Laboratory, Charles Perkins Centre, Faculty of Medicine and HealthUniversity of SydneySydneyNSWAustralia
| | - Jean Y Yang
- School of Mathematics and StatisticsUniversity of SydneySydneyNSWAustralia
- Charles Perkins CentreUniversity of SydneySydneyNSWAustralia
| | - Barbara Fazekas de St Groth
- Charles Perkins CentreUniversity of SydneySydneyNSWAustralia
- School of Medical Sciences, Faculty of Medicine and HealthUniversity of SydneySydneyNSWAustralia
- Ramaciotti Facility for Human Systems BiologyUniversity of SydneySydneyNSWAustralia
| | - Helen M McGuire
- Charles Perkins CentreUniversity of SydneySydneyNSWAustralia
- School of Medical Sciences, Faculty of Medicine and HealthUniversity of SydneySydneyNSWAustralia
- Ramaciotti Facility for Human Systems BiologyUniversity of SydneySydneyNSWAustralia
| | - Gemma A Figtree
- Cardiothoracic and Vascular HealthKolling Institute of Medical ResearchSydneyNSWAustralia
- Department of Cardiology, Royal North Shore HospitalNorthern Sydney Local Health DistrictSydneyNSWAustralia
- Northern Clinical School, Faculty of Medicine and HealthUniversity of SydneySydneyNSWAustralia
- Charles Perkins CentreUniversity of SydneySydneyNSWAustralia
| |
Collapse
|
3
|
Huang J, Wang F, Tang X. Uncovering the shared molecule and mechanism between ulcerative colitis and atherosclerosis: an integrative genomic analysis. Front Immunol 2023; 14:1219457. [PMID: 37638002 PMCID: PMC10450151 DOI: 10.3389/fimmu.2023.1219457] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Accepted: 07/24/2023] [Indexed: 08/29/2023] Open
Abstract
Background Ulcerative colitis (UC) and atherosclerosis (AS) are closely related. However, the pathologic mechanisms underlying the co-occurrence of UC and AS are not well understood. Objects To reveal the hub molecule and mechanism involved in the co-occurrence of UC and AS. Methods Differentially expressed genes (DEGs) of UC and AS were obtained, and the shared DEGs of UC and AS were explored for biological function. Next, the hub genes were explored using the cytoHubba plugin. The predictive ability of the hub genes was measured by constructing the receiver operating characteristic curve. Analyses of immune infiltration and the single-gene gene set enrichment analysis (GSEA) for the hub genes were further carried out. Results Identification of 59 DEGs (55 were upregulated and four were downregulated) shared by both UC and AS was performed. Enriched pathways of the shared DEGs were mainly related to immunity and inflammation. Protein tyrosine phosphatase, receptor type, C (PTPRC) was identified as the hub crosstalk gene for the comorbidity of UC and AS. The upregulation of PTPRC was correlated with mast cells resting, T cells CD4 memory resting, macrophages M0, and macrophages M1. Pathways of immune and inflammatory processes, including NF-kappa B, viral protein interaction with cytokine and cytokine receptor, and cytokine-cytokine receptor interaction, were significantly correlated with high expression of PTPRC in UC and AS. Conclusion At the transcriptional level, our study reveals that imbalanced inflammatory and immune responses are the key pathological mechanisms underlying the comorbidity of UC and AS and that PTPRC is a key biomarker for the comorbidity of UC and AS.
Collapse
Affiliation(s)
- Jinke Huang
- Department of Gastroenterology, Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing, China
- Institute of Digestive Diseases, Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing, China
| | - Fengyun Wang
- Department of Gastroenterology, Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing, China
- Institute of Digestive Diseases, Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing, China
| | - Xudong Tang
- Institute of Digestive Diseases, Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing, China
| |
Collapse
|
4
|
Nambo-Venegas R, Palacios-González B, Mas-Oliva J, Aurioles-Amozurrutia AK, Cruz-Rangel A, Moreno A, Hidalgo-Miranda A, Rodríguez-Dorantes M, Vadillo-Ortega F, Xicohtencatl-Cortes J, Ruiz-Olmedo MI, Reyes-Grajeda JP. Conversion of M1 Macrophages to Foam Cells: Transcriptome Differences Determined by Sex. Biomedicines 2023; 11:biomedicines11020490. [PMID: 36831031 PMCID: PMC9953229 DOI: 10.3390/biomedicines11020490] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Revised: 01/27/2023] [Accepted: 01/30/2023] [Indexed: 02/11/2023] Open
Abstract
BACKGROUND M1 macrophages involved in pro-inflammatory processes can be induced by low-density lipoproteins (LDL), giving rise to foam cells. In the atheroma plaque, it has been identified that males present more advanced lesions associated with infiltration. Therefore, our study aims to investigate sex-related changes in the transcriptome of M1 macrophages during the internalization process of LDL particles. METHODS Peripheral blood mononuclear cells (PBMCs) from healthy male and female subjects were separated using Hystopaque, and monocytes were isolated from PBMCs using a positive selection of CD14+ cells. Cells were stimulated with LDL 10 µg/mL, and the transcriptional profile of M1 macrophages performed during LDL internalization was determined using a Clariom D platform array. RESULTS Chromosome Y influences the immune system and inflammatory responses in males expressing 43% of transcripts in response to LDL treatment. Males and females share 15 transcripts, where most correspond to non-coding elements involved in oxidative stress and endothelial damage. CONCLUSIONS During LDL internalization, male monocyte-derived M1 macrophages display more marked proinflammatory gene expression. In contrast, female M1 macrophages display a more significant number of markers associated with cell damage.
Collapse
Affiliation(s)
- Rafael Nambo-Venegas
- Laboratorio de Estructura de Proteínas, Instituto Nacional de Medicina Genómica, Mexico City 14600, Mexico
| | - Berenice Palacios-González
- Laboratorio de Envejecimiento Saludable, Centro de Investigación Sobre Envejecimiento (CIE-CINVESTAV Sur), Instituto Nacional de Medicina Genómica, Mexico City 14330, Mexico
| | - Jaime Mas-Oliva
- Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Mexico City 04510, Mexico
| | | | - Armando Cruz-Rangel
- Laboratorio de Estructura de Proteínas, Instituto Nacional de Medicina Genómica, Mexico City 14600, Mexico
| | - Abel Moreno
- Instituto de Química, Universidad Nacional Autónoma de México, Mexico City 04510, Mexico
| | - Alfredo Hidalgo-Miranda
- Laboratorio de Genómica del Cáncer, Instituto Nacional de Medicina Genómica, Mexico City 14600, Mexico
| | | | - Felipe Vadillo-Ortega
- Unidad de Vinculación Científica de la Facultad de Medicina UNAM en INMEGEN, Instituto Nacional de Medicina Genómica, Mexico City 14600, Mexico
| | - Juan Xicohtencatl-Cortes
- Laboratorio de Bacteriología Intestinal, Hospital Infantil de México Federico Gómez, Mexico City 06720, Mexico
| | | | - Juan Pablo Reyes-Grajeda
- Laboratorio de Estructura de Proteínas, Instituto Nacional de Medicina Genómica, Mexico City 14600, Mexico
- Correspondence:
| |
Collapse
|
5
|
Paunel-Görgülü A, Conforti A, Mierau N, Zierden M, Xiong X, Wahlers T. Peptidylarginine deiminase 4 deficiency in bone marrow cells prevents plaque progression without decreasing atherogenic inflammation in apolipoprotein E-knockout mice. Front Cardiovasc Med 2022; 9:1046273. [PMID: 36465436 PMCID: PMC9709396 DOI: 10.3389/fcvm.2022.1046273] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Accepted: 11/01/2022] [Indexed: 08/30/2023] Open
Abstract
INTRODUCTION Despite multiple studies in the past, the role of peptidylarginine deiminase 4 (PAD4) in atherosclerosis is currently insufficiently understood. In this regard, PAD4 deletion or inhibition of enzymatic activity was previously reported to ameliorate disease progression and inflammation. Besides, strong influence of neutrophil extracellular traps (NETs) on atherosclerosis burden has been proposed. Here, we studied the role of PAD4 for atherogenesis and plaque progression in a mouse model of atherosclerosis. METHODS AND RESULTS Lethally irradiated ApoE -/- mice were reconstituted with ApoE -/-/Pad4 -/- bone marrow cells and fed a high-fat diet (HFD) for 4 and 10 weeks, respectively. PAD4 deficiency did not prevent the development of atherosclerotic lesions after 4 weeks of HFD. However, after 10 weeks of HFD, mice with bone marrow cells-restricted PAD4 deficiency displayed significantly reduced lesion size, impaired lipid incorporation, decreased necrotic core area and less collagen when compared to ApoE -/- bone marrow-transplanted mice as demonstrated by histological staining. Moreover, flow cytometric analysis and quantitative real-time PCR revealed different macrophage subsets in atherosclerotic lesions and higher inflammatory response in these mice, as reflected by increased content of M1-like macrophages and upregulated aortic expression of the pro-inflammatory genes CCL2 and iNOS. Notably, diminished oxLDL uptake by in vitro-polarized M1-like macrophages was evidenced when compared to M2-like cells. CONCLUSION These results suggest that pharmacological inhibition of PAD4 may impede lipid accumulation and lesion progression despite no beneficial effects on vascular inflammation.
Collapse
Affiliation(s)
- Adnana Paunel-Görgülü
- Department of Cardiothoracic Surgery, Heart Center, University of Cologne, Cologne, Germany
| | - Andreas Conforti
- Department of Cardiothoracic Surgery, Heart Center, University of Cologne, Cologne, Germany
| | - Natalia Mierau
- Department of Cardiothoracic Surgery, Heart Center, University of Cologne, Cologne, Germany
| | - Mario Zierden
- Department of Cardiology, Heart Center, University of Cologne, Cologne, Germany
- Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany
| | - Xiaolin Xiong
- Department of Cardiothoracic Surgery, Heart Center, University of Cologne, Cologne, Germany
| | - Thorsten Wahlers
- Department of Cardiothoracic Surgery, Heart Center, University of Cologne, Cologne, Germany
| |
Collapse
|
6
|
Vellasamy DM, Lee SJ, Goh KW, Goh BH, Tang YQ, Ming LC, Yap WH. Targeting Immune Senescence in Atherosclerosis. Int J Mol Sci 2022; 23:13059. [PMID: 36361845 PMCID: PMC9658319 DOI: 10.3390/ijms232113059] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 10/07/2022] [Accepted: 10/10/2022] [Indexed: 10/29/2023] Open
Abstract
Atherosclerosis is one of the main underlying causes of cardiovascular diseases (CVD). It is associated with chronic inflammation and intimal thickening as well as the involvement of multiple cell types including immune cells. The engagement of innate or adaptive immune response has either athero-protective or atherogenic properties in exacerbating or alleviating atherosclerosis. In atherosclerosis, the mechanism of action of immune cells, particularly monocytes, macrophages, dendritic cells, and B- and T-lymphocytes have been discussed. Immuno-senescence is associated with aging, viral infections, genetic predispositions, and hyperlipidemia, which contribute to atherosclerosis. Immune senescent cells secrete SASP that delays or accelerates atherosclerosis plaque growth and associated pathologies such as aneurysms and coronary artery disease. Senescent cells undergo cell cycle arrest, morphological changes, and phenotypic changes in terms of their abundances and secretome profile including cytokines, chemokines, matrix metalloproteases (MMPs) and Toll-like receptors (TLRs) expressions. The senescence markers are used in therapeutics and currently, senolytics represent one of the emerging treatments where specific targets and clearance of senescent cells are being considered as therapy targets for the prevention or treatment of atherosclerosis.
Collapse
Affiliation(s)
- Danusha Michelle Vellasamy
- School of Biosciences, Faculty of Medical and Health Sciences, Taylor’s University, Subang Jaya 47500, Malaysia
| | - Sin-Jye Lee
- School of Biosciences, Faculty of Medical and Health Sciences, Taylor’s University, Subang Jaya 47500, Malaysia
| | - Khang Wen Goh
- Faculty of Data Science and Information Technology, INTI International University, Nilai 71800, Malaysia
| | - Bey-Hing Goh
- Biofunctional Molecule Exploratory (BMEX) Research Group, School of Pharmacy, Monash University Malaysia, Bandar Sunway 47500, Malaysia
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou 310058, China
| | - Yin-Quan Tang
- School of Biosciences, Faculty of Medical and Health Sciences, Taylor’s University, Subang Jaya 47500, Malaysia
- Centre for Drug Discovery and Molecular Pharmacology, Faculty of Medical and Health Sciences, Taylor’s University, Subang Jaya 47500, Malaysia
| | - Long Chiau Ming
- PAP Rashidah Sa’adatul Bolkiah Institute of Health Sciences, Universiti Brunei Darussalam, Gadong BE1410, Brunei
| | - Wei Hsum Yap
- School of Biosciences, Faculty of Medical and Health Sciences, Taylor’s University, Subang Jaya 47500, Malaysia
- Centre for Drug Discovery and Molecular Pharmacology, Faculty of Medical and Health Sciences, Taylor’s University, Subang Jaya 47500, Malaysia
| |
Collapse
|
7
|
Krga I, Corral-Jara KF, Barber-Chamoux N, Dubray C, Morand C, Milenkovic D. Grapefruit Juice Flavanones Modulate the Expression of Genes Regulating Inflammation, Cell Interactions and Vascular Function in Peripheral Blood Mononuclear Cells of Postmenopausal Women. Front Nutr 2022; 9:907595. [PMID: 35694160 PMCID: PMC9178201 DOI: 10.3389/fnut.2022.907595] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Accepted: 04/26/2022] [Indexed: 12/04/2022] Open
Abstract
Grapefruit is a rich source of flavanones, phytochemicals suggested excreting vasculoprotective effects. We previously showed that flavanones in grapefruit juice (GFJ) reduced postmenopausal women’s pulse-wave velocity (PWV), a measure of arterial stiffness. However, mechanisms of flavanone action in humans are largely unknown. This study aimed to decipher molecular mechanisms of flavanones by multi-omics analysis in PBMCs of volunteers consuming GFJ and flavanone-free control drink for 6 months. Modulated genes and microRNAs (miRNAs) were identified using microarrays. Bioinformatics analyses assessed their functions, interactions and correlations with previously observed changes in PWV. GFJ modified gene and miRNA expressions. Integrated analysis of modulated genes and miRNA-target genes suggests regulation of inflammation, immune response, cell interaction and mobility. Bioinformatics identified putative mediators of the observed nutrigenomic effect (STAT3, NF-κB) and molecular docking demonstrated potential binding of flavanone metabolites to transcription factors and cell-signaling proteins. We also observed 34 significant correlations between changes in gene expression and PWV. Moreover, global gene expression was negatively correlated with gene expression profiles in arterial stiffness and hypertension. This study revealed molecular mechanisms underlying vasculoprotective effects of flavanones, including interactions with transcription factors and gene and miRNA expression changes that inversely correlate with gene expression profiles associated with cardiovascular risk factors.
Collapse
Affiliation(s)
- Irena Krga
- Centre of Research Excellence in Nutrition and Metabolism, Institute for Medical Research, National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | | | | | - Claude Dubray
- Institut National de la Santé et de la Recherche Médicale (INSERM), CIC 501, UMR 766, Clermont-Ferrand, France
| | - Christine Morand
- Université Clermont Auvergne, INRAE, UNH, Clermont-Ferrand, France
| | - Dragan Milenkovic
- Université Clermont Auvergne, INRAE, UNH, Clermont-Ferrand, France
- Department of Nutrition, College of Agricultural and Environmental Sciences, University of California, Davis, Davis, CA, United States
- *Correspondence: Dragan Milenkovic,
| |
Collapse
|
8
|
Madonna R. Angiocrine endothelium: From physiology to atherosclerosis and cardiac repair. Vascul Pharmacol 2022; 144:106993. [DOI: 10.1016/j.vph.2022.106993] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Revised: 03/25/2022] [Accepted: 03/30/2022] [Indexed: 02/08/2023]
|
9
|
Meng Z, Hernandez R, Liu J, Gwag T, Lu W, Hsiai TK, Kaul M, Zhou T, Zhou C. HIV Protein Tat Induces Macrophage Dysfunction and Atherosclerosis Development in Low-Density Lipoprotein Receptor-Deficient Mice. Cardiovasc Drugs Ther 2022; 36:201-215. [PMID: 33459922 PMCID: PMC8286272 DOI: 10.1007/s10557-021-07141-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 01/06/2021] [Indexed: 12/17/2022]
Abstract
PURPOSE HIV infection is consistently associated with an increased risk of atherosclerotic cardiovascular disease, but the underlying mechanisms remain elusive. HIV protein Tat, a transcriptional activator of HIV, has been shown to activate NF-κB signaling and promote inflammation in vitro. However, the atherogenic effects of HIV Tat have not been investigated in vivo. Macrophages are one of the major cell types involved in the initiation and progression of atherosclerosis. We and others have previously revealed the important role of IκB kinase β (IKKβ), a central inflammatory coordinator through activating NF-κB, in the regulation of macrophage functions and atherogenesis. This study investigated the impact of HIV Tat exposure on macrophage functions and atherogenesis. METHODS To investigate the effects of Tat on macrophage IKKβ activation and atherosclerosis development in vivo, myeloid-specific IKKβ-deficient LDLR-deficient (IKKβΔMyeLDLR-/-) mice and their control littermates (IKKβF/FLDLR-/-) were exposed to recombinant HIV protein Tat. RESULTS Exposure to Tat significantly increased atherosclerotic lesion size and plaque vulnerability in IKKβF/FLDLR-/- but not IKKβΔMyeLDLR-/- mice. Deficiency of myeloid IKKβ attenuated Tat-elicited macrophage inflammatory responses and atherosclerotic lesional inflammation in IKKβΔMyeLDLR-/- mice. Further, RNAseq analysis demonstrated that HIV protein Tat affects the expression of many atherosclerosis-related genes in vitro in an IKKβ-dependent manner. CONCLUSIONS Our findings reveal atherogenic effects of HIV protein Tat in vivo and demonstrate a pivotal role of myeloid IKKβ in Tat-driven atherogenesis.
Collapse
Affiliation(s)
- Zhaojie Meng
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, CA, USA
| | - Rebecca Hernandez
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, CA, USA
| | - Jingwei Liu
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, CA, USA
| | - Taesik Gwag
- Department of Pharmacology and Nutritional Sciences, College of Medicine, University of Kentucky, Lexington, KY, USA
| | - Weiwei Lu
- Department of Pharmacology and Nutritional Sciences, College of Medicine, University of Kentucky, Lexington, KY, USA
| | - Tzung K Hsiai
- Departments of Medicine and Bioengineering, David Geffen School of Medicine, Henry Samueli School of Engineering and Applied Science, University of California, Los Angeles, CA, USA
| | - Marcus Kaul
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, CA, USA
| | - Tong Zhou
- Department of Physiology and Cell Biology, Reno School of Medicine, University of Nevada, Reno, NV, USA
| | - Changcheng Zhou
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, CA, USA.
| |
Collapse
|
10
|
Pakzad B, Rajae E, Shahrabi S, Mansournezhad S, Davari N, Azizidoost S, Saki N. T-Cell Molecular Modulation Responses in Atherosclerosis Anergy. Lab Med 2021; 51:557-565. [PMID: 32106301 DOI: 10.1093/labmed/lmaa003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Atherosclerosis continues to be a major cause of death in patients with cardiovascular diseases. The cooperative role of immunity has been recently considered in atherosclerotic plaque inflammation, especially adaptive immune response by T cells. In this review, we examine the possible role of T cells in atherosclerosis-mediated inflammation and conceivable therapeutic strategies that can ameliorate complications of atherosclerosis. The cytokines secreted by T-lymphocyte subsets, different pathophysiological profiles of microRNAs (miRs), and the growth factor/receptor axis have diverse effects on the inflammatory cycle of atherosclerosis. Manipulation of miRNA expression and prominent growth factor receptors involved in inflammatory cytokine secretion in atherosclerosis can be considered diagnostic biomarkers in the induction of anergy and blockade of atherosclerotic development. This manuscript reviews immunomodulation of T cells responses in atherosclerosis anergy.
Collapse
Affiliation(s)
- Bahram Pakzad
- Internal Medicine Department, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Elham Rajae
- Department of Rheumatology, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Saeid Shahrabi
- -Department of Biochemistry and Hematology, Faculty of Medicine, Semnan University of Medical Sciences, Semnan, Iran
| | - Somayeh Mansournezhad
- Thalassemia and Hemoglobinopathy Research Center, Research Institute of Health, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Nader Davari
- Thalassemia and Hemoglobinopathy Research Center, Research Institute of Health, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Shirin Azizidoost
- Thalassemia and Hemoglobinopathy Research Center, Research Institute of Health, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Najmaldin Saki
- Thalassemia and Hemoglobinopathy Research Center, Research Institute of Health, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| |
Collapse
|
11
|
Mahmoudi E, Mozhgani SH, Sharifinejad N. The role of mycobiota-genotype association in inflammatory bowel diseases: a narrative review. Gut Pathog 2021; 13:31. [PMID: 33964975 PMCID: PMC8106830 DOI: 10.1186/s13099-021-00426-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/24/2020] [Accepted: 04/29/2021] [Indexed: 02/06/2023] Open
Abstract
Inflammatory bowel disease (IBD) is a chronic inflammatory disease affecting various parts of the gastrointestinal tract. A majority of the current evidence points out the involvement of intestinal dysbiosis in the IBD pathogenesis. Recently, the association of intestinal fungal composition With IBD susceptibility and severity has been reported. These studies suggested gene polymorphisms in the front line of host defense against intestinal microorganisms are considered to play a role in IBD pathogenesis. The studies have also detected increased susceptibility to fungal infections in patients carrying IBD-related mutations. Therefore, a literature search was conducted in related databases to review articles addressing the mycobiota-genotype association in IBD.
Collapse
Affiliation(s)
- Elaheh Mahmoudi
- Division of Mycology, School of Medicine, Alborz University of Medical Sciences, Karaj, Iran
| | - Sayed-Hamidreza Mozhgani
- Department of Microbiology, School of Medicine, Alborz University of Medical Sciences, Karaj, Iran
| | - Niusha Sharifinejad
- Student Research Committee, Alborz University of Medical Sciences, Karaj, Iran. .,Alborz Office of USERN, Universal Scientific Education and Research Network (USERN), Alborz University of Medical Sciences, Karaj, Iran.
| |
Collapse
|
12
|
Bigford GE, Szeto A, Kimball J, Herderick EE, Mendez AJ, Nash MS. Cardiometabolic risks and atherosclerotic disease in ApoE knockout mice: Effect of spinal cord injury and Salsalate anti-inflammatory pharmacotherapy. PLoS One 2021; 16:e0246601. [PMID: 33626069 PMCID: PMC7904230 DOI: 10.1371/journal.pone.0246601] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Accepted: 01/21/2021] [Indexed: 12/05/2022] Open
Abstract
OBJECTIVE To test in mice with a double mutation of the ApoE gene (ApoE-/-) whether spinal cord injury (SCI) hastens the native trajectory of, and established component risks for, atherosclerotic disease (AD), and whether Salsalate anti-inflammatory pharmacotherapy attenuates the impact of SCI. METHODS ApoE-/- mice were anesthetized and underwent a T9 laminectomy. Exposed spinal cords were given a contusion injury (70 k-dynes). Sham animals underwent all surgical procedures, excluding injury. Injured animals were randomized to 2 groups: SCI or SCI+Salsalate [120 mg/Kg/day i.p.]. Mice were serially sacrificed at 20-, 24-, and 28-weeks post-SCI, and body mass was recorded. At sacrifice, heart and aorta were harvested intact, fixed in 10% buffered formalin, cleaned and cut longitudinally for en face preparation. The aortic tree was stained with oil-red-O (ORO). AD lesion histomorphometry was calculated from the proportional area of ORO. Plasma total cholesterol, triglycerides and proatherogenic inflammatory cytokines (PAIC's) were analyzed. RESULTS AD lesion in the aortic arch progressively increased in ApoE-/-, significant at 24- and 28-weeks. AD in SCI is significantly greater at 24- and 28-weeks compared to time-controlled ApoE-/-. Salsalate treatment attenuates the SCI-induced increase at these time points. Body mass in all SCI groups are significantly reduced compared to time-controlled ApoE-/-. Cholesterol and triglycerides are significantly higher with SCI by 24- and 28-weeks, compared to ApoE-/-, and Salsalate reduces the SCI-induced effect on cholesterol. PAIC's interleukin-1β (IL-1β), interleukin-6 (IL-6), tumor necrosis factor α (TNFα), monocyte chemoattractant protein-1 (MCP-1), and chemokine (C-C motif) ligand 5 (CCL-5) are significantly greater with SCI compared to ApoE-/- at varying timepoints. Salsalate confers a marginal reducing effect on PAIC's by 28-weeks compared to SCI. Regression models determine that each PAIC is a significant and positive predictor of lesion. (p's <0.05). CONCLUSIONS SCI accelerates aortic AD and associated risk factors, and anti-inflammatory treatment may attenuate the impact of SCI on AD outcomes. PAIC's IL-1β, IL-6, TNFα, MCP-1, and CCL-5 may be effective predictors of AD.
Collapse
Affiliation(s)
- Gregory E. Bigford
- Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, Florida, United States of America
| | - Angela Szeto
- Department of Medicine, University of Miami Miller School of Medicine, Miami, Florida, United States of America
| | - John Kimball
- Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, Florida, United States of America
| | | | - Armando J. Mendez
- Department of Medicine, University of Miami Miller School of Medicine, Miami, Florida, United States of America
| | - Mark S. Nash
- Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, Florida, United States of America
- Department of Physical Medicine and Rehabilitation, University of Miami Miller School of Medicine, Miami, Florida, United States of America
- Department of Physical Therapy, University of Miami, Coral Gables, Florida, United States of America
| |
Collapse
|
13
|
Li J, Peng L, Bai W, Peng P, Chen W, Yang W, Shao J. Biliverdin Protects Against Cerebral Ischemia/Reperfusion Injury by Regulating the miR-27a-3p/Rgs1 Axis. Neuropsychiatr Dis Treat 2021; 17:1165-1181. [PMID: 33911865 PMCID: PMC8075361 DOI: 10.2147/ndt.s300773] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Accepted: 04/02/2021] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND We have previously demonstrated that biliverdin has neuroprotective effects that ameliorate cerebral ischemia/reperfusion (I/R) injury in rats. However, the underlying mechanism is unknown. This study aimed at elucidating on the modulatory role of miR-27a-3p on Rgs1 as a mechanism by which biliverdin affects cerebral I/R injury. METHODS Middle cerebral artery occlusion/reperfusion (MCAO/R) was used to establish I/R rat models while oxygen glucose deprivation/reoxygenation (OGD/R) was used to induce hippocampal neurons to establish I/R models in vitro. Infarct volume was assessed by TTC staining. Apoptotic analyses of ischemic cortical neurons and cells were performed by TUNEL staining and flow cytometry, respectively. Cell viability was assessed by the CCK-8 assay while the target of miR-27a-3p was determined by double luciferase reporter assay. Relative expression levels of miR-27a-3p and Rgs1 (in vivo and in vitro) as well as markers of inflammation and apoptosis (in vitro) were detected by RT-qPCR. Then, Elisa and western blot were used to assess protein expression levels of inflammatory and apoptotic markers in vitro. RESULTS Biliverdin suppressed inflammation and apoptosis in hippocampal neurons upon OGD/R, and reduced cerebral infarction volume as well as apoptosis in the MCAO/R rat model. Furthermore, biliverdin upregulated miR-27a-3p and downregulated hippocampal neuron Rgs1 after OGD/R as well as in rat brain tissues after cerebral I/R. Bioinformatic analysis revealed an miR-27a-3p docking site in the 3'-UTR region of Rgs1. Luciferase reporter assays showed that Rgs1 is an miR-27a-3p target. Moreover, miR-27a-3p upregulation inhibited OGD/R-triggered inflammation and suppressed neuronal apoptosis. Rgs1 knockdown suppressed OGD/R-triggered inflammation and decreased neuronal apoptosis while miR-27a-3p downregulation reversed the protective effect of Rgs1 knockdown. Moreover, miR-27a-3p overexpression and Rgs1 silencing suppressed NF-κB (p65) expression. CONCLUSION Biliverdin protects against cerebral I/R injury by regulating the miR-27a-3p/Rgs1 axis, thereby inhibiting inflammation and apoptosis.
Collapse
Affiliation(s)
- Junjie Li
- Department of Anesthesiology, First Affiliated Hospital, Kunming Medical University, Kunming City, 650032, People's Republic of China
| | - Lijia Peng
- Department of Anesthesiology, First Affiliated Hospital, Kunming Medical University, Kunming City, 650032, People's Republic of China
| | - Wenya Bai
- Department of Anesthesiology, First Affiliated Hospital, Kunming Medical University, Kunming City, 650032, People's Republic of China
| | - Peihua Peng
- Department of Anesthesiology, First Affiliated Hospital, Kunming Medical University, Kunming City, 650032, People's Republic of China
| | - Wendong Chen
- Department of Anesthesiology, First Affiliated Hospital, Kunming Medical University, Kunming City, 650032, People's Republic of China
| | - Wei Yang
- Department of Anesthesiology, First Affiliated Hospital, Kunming Medical University, Kunming City, 650032, People's Republic of China
| | - Jianlin Shao
- Department of Anesthesiology, First Affiliated Hospital, Kunming Medical University, Kunming City, 650032, People's Republic of China
| |
Collapse
|
14
|
Brandão SCS, Godoi ETAM, Ramos JDOX, de Melo LMMP, Sarinho ESC. Severe COVID-19: understanding the role of immunity, endothelium, and coagulation in clinical practice. J Vasc Bras 2020; 19:e20200131. [PMID: 34211530 PMCID: PMC8218014 DOI: 10.1590/1677-5449.200131] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Accepted: 08/17/2020] [Indexed: 01/08/2023] Open
Abstract
SARS-CoV-2 is responsible for the COVID-19 pandemic. The immune system is a determinant factor in defense against viral infections. Thus, when it acts in a balanced and effective manner the disease is self-limited and benign. Nevertheless, in a significant proportion of the population, the immune response is exaggerated. When infected, patients with diabetes, hypertension, obesity, and cardiovascular disease are more likely to progress to severe forms. These diseases are related to chronic inflammation and endothelial dysfunction. Toll-like receptors are expressed on immune cells and play an important role in the physiopathology of cardiovascular and metabolic diseases. When activated, they can induce release of inflammatory cytokines. Hypercoagulability, hyperinflammation, platelet hyperresponsiveness, and endothelial dysfunction occur in immune system hyperactivity caused by viral activity, thereby increasing the risk of arterial and venous thrombosis. We discuss the interactions between COVID-19, immunity, the endothelium, and coagulation, as well as why cardiometabolic diseases have a negative impact on COVID-19 prognosis.
Collapse
|
15
|
Wellmann IAM, Ibiapina HNS, Sachett JAG, Sartim MA, Silva IM, Oliveira SS, Tarragô AM, Moura-da-Silva AM, Lacerda MVG, Ferreira LCDL, Malheiro A, Monteiro WM, Costa AG. Correlating Fibrinogen Consumption and Profiles of Inflammatory Molecules in Human Envenomation's by Bothrops atrox in the Brazilian Amazon. Front Immunol 2020; 11:1874. [PMID: 32973773 PMCID: PMC7468254 DOI: 10.3389/fimmu.2020.01874] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Accepted: 07/13/2020] [Indexed: 12/11/2022] Open
Abstract
Snakebites are considered a major public health problem worldwide. In the Amazon region of Brazil, the snake Bothrops atrox (B. atrox) is responsible for 90% of the bites. These bites may cause local and systemic signs from acute inflammatory reaction and hemostatic changes, and present common hemorrhagic disorders. These alterations occur due the action of hemostatically active and immunogenic toxins which are capable of triggering a wide range of hemostatic and inflammatory events. However, the crosstalk between coagulation disorders and inflammatory reaction still has gaps in snakebites. Thus, the goal of this study was to describe the relationship between the consumption of fibrinogen and the profile of inflammatory molecules (chemokines and cytokines) in evenomations by B. atrox snakebites. A prospective study was carried out with individuals who had suffered B. atrox snakebites and presented different levels of fibrinogen consumption (normal fibrinogen [NF] and hypofibrinogenemia [HF]). Seventeen patients with NF and 55 patients with HF were eligible for the study, in addition to 50 healthy controls (CG). The molecules CXCL-8, CCL-5, CXCL-9, CCL-2, CXCL-10, IL-6, TNF, IL-2, IL-10, IFN-γ, IL-4, and IL-17A were quantified in plasma using the CBA technique at three different times (pre-antivenom therapy [T0], 24 h [T1], and 48 h [T2] after antivenom therapy). The profile of the circulating inflammatory response is different between the groups studied, with HF patients having higher concentrations of CCL-5 and lower IFN-γ. In addition, antivenom therapy seems to have a positive effect, leading to a profile of circulating inflammatory response similar in quantification of T1 and T2 on both groups. Furthermore, these results suggest that a number of interactions of CXCL-8, CXCL-9, CCL-2, IL-6, and IFN-γ in HF patients are directly affected by fibrinogen levels, which may be related to the inflammatory response and coagulation mutual relationship induced by B. atrox venom. The present study is the first report on inflammation-coagulation crosstalk involving snakebite patients and supports the better understanding of envenomation's pathophysiology mechanisms and guides in the search for novel biomarkers and prospective therapies.
Collapse
Affiliation(s)
- Irmgardt Alicia María Wellmann
- Programa de Pós-Graduação em Medicina Tropical, Universidade do Estado do Amazonas (UEA), Manaus, Brazil.,Instituto de Pesquisa Clínica Carlos Borborema (IPCCB), Fundação de Medicina Tropical Dr. Heitor Vieira Dourado (FMT-HVD), Manaus, Brazil
| | - Hiochelson Najibe Santos Ibiapina
- Programa de Pós-Graduação em Medicina Tropical, Universidade do Estado do Amazonas (UEA), Manaus, Brazil.,Instituto de Pesquisa Clínica Carlos Borborema (IPCCB), Fundação de Medicina Tropical Dr. Heitor Vieira Dourado (FMT-HVD), Manaus, Brazil
| | - Jacqueline Almeida Gonçalves Sachett
- Programa de Pós-Graduação em Medicina Tropical, Universidade do Estado do Amazonas (UEA), Manaus, Brazil.,Instituto de Pesquisa Clínica Carlos Borborema (IPCCB), Fundação de Medicina Tropical Dr. Heitor Vieira Dourado (FMT-HVD), Manaus, Brazil
| | - Marco Aurélio Sartim
- Programa de Pós-Graduação em Imunologia Básica e Aplicada, Universidade Federal do Amazonas (UFAM), Manaus, Brazil
| | - Iran Mendonça Silva
- Instituto de Pesquisa Clínica Carlos Borborema (IPCCB), Fundação de Medicina Tropical Dr. Heitor Vieira Dourado (FMT-HVD), Manaus, Brazil
| | - Sâmella Silva Oliveira
- Programa de Pós-Graduação em Medicina Tropical, Universidade do Estado do Amazonas (UEA), Manaus, Brazil.,Instituto de Pesquisa Clínica Carlos Borborema (IPCCB), Fundação de Medicina Tropical Dr. Heitor Vieira Dourado (FMT-HVD), Manaus, Brazil
| | - Andréa Monteiro Tarragô
- Programa de Pós-Graduação em Imunologia Básica e Aplicada, Universidade Federal do Amazonas (UFAM), Manaus, Brazil.,Programa de Pós-Graduação em Ciências Aplicadas à Hematologia, Universidade do Estado do Amazonas (UEA), Manaus, Brazil.,Diretoria de Ensino e Pesquisa, Fundação Hospitalar de Hematologia e Hemoterapia do Amazonas (HEMOAM), Manaus, Brazil
| | - Ana Maria Moura-da-Silva
- Programa de Pós-Graduação em Medicina Tropical, Universidade do Estado do Amazonas (UEA), Manaus, Brazil.,Laboratório de Imunopatologia, Instituto Butantan, São Paulo, Brazil
| | - Marcus Vinícius Guimarães Lacerda
- Programa de Pós-Graduação em Medicina Tropical, Universidade do Estado do Amazonas (UEA), Manaus, Brazil.,Instituto de Pesquisa Clínica Carlos Borborema (IPCCB), Fundação de Medicina Tropical Dr. Heitor Vieira Dourado (FMT-HVD), Manaus, Brazil.,Programa de Pós-Graduação em Ciências Aplicadas à Hematologia, Universidade do Estado do Amazonas (UEA), Manaus, Brazil.,Instituto de Pesquisas Leônidas e Maria Deane, FIOCRUZ-Amazônia, Manaus, Brazil
| | - Luiz Carlos de Lima Ferreira
- Programa de Pós-Graduação em Medicina Tropical, Universidade do Estado do Amazonas (UEA), Manaus, Brazil.,Instituto de Pesquisa Clínica Carlos Borborema (IPCCB), Fundação de Medicina Tropical Dr. Heitor Vieira Dourado (FMT-HVD), Manaus, Brazil
| | - Adriana Malheiro
- Programa de Pós-Graduação em Medicina Tropical, Universidade do Estado do Amazonas (UEA), Manaus, Brazil.,Programa de Pós-Graduação em Imunologia Básica e Aplicada, Universidade Federal do Amazonas (UFAM), Manaus, Brazil.,Programa de Pós-Graduação em Ciências Aplicadas à Hematologia, Universidade do Estado do Amazonas (UEA), Manaus, Brazil.,Diretoria de Ensino e Pesquisa, Fundação Hospitalar de Hematologia e Hemoterapia do Amazonas (HEMOAM), Manaus, Brazil
| | - Wuelton Marcelo Monteiro
- Programa de Pós-Graduação em Medicina Tropical, Universidade do Estado do Amazonas (UEA), Manaus, Brazil.,Instituto de Pesquisa Clínica Carlos Borborema (IPCCB), Fundação de Medicina Tropical Dr. Heitor Vieira Dourado (FMT-HVD), Manaus, Brazil
| | - Allyson Guimarães Costa
- Programa de Pós-Graduação em Medicina Tropical, Universidade do Estado do Amazonas (UEA), Manaus, Brazil.,Instituto de Pesquisa Clínica Carlos Borborema (IPCCB), Fundação de Medicina Tropical Dr. Heitor Vieira Dourado (FMT-HVD), Manaus, Brazil.,Programa de Pós-Graduação em Imunologia Básica e Aplicada, Universidade Federal do Amazonas (UFAM), Manaus, Brazil.,Programa de Pós-Graduação em Ciências Aplicadas à Hematologia, Universidade do Estado do Amazonas (UEA), Manaus, Brazil.,Diretoria de Ensino e Pesquisa, Fundação Hospitalar de Hematologia e Hemoterapia do Amazonas (HEMOAM), Manaus, Brazil
| |
Collapse
|
16
|
Wu Y, Xu W, Hou J, Liu Y, Li R, Liu J, Li C, Tang X, Lin L, Pan Y, Zhang D. Porphyromonas gingivalis-Induced MIF Regulates Intercellular Adhesion Molecule-1 Expression in EA.hy926 Cells and Monocyte-Endothelial Cell Adhesion Through the Receptors CD74 and CXCR4. Inflammation 2019; 42:874-883. [PMID: 30506423 PMCID: PMC6527533 DOI: 10.1007/s10753-018-0942-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Porphyromonas gingivalis (P. gingivalis) is an important pathogen that contributes to periodontal disease and causes infections that promote the progression of atherosclerosis. Our previous studies showed that macrophage migration inhibitory factor (MIF) facilitates monocyte adhesion to endothelial cells by regulating the expression of intercellular adhesion molecule-1 (ICAM-1) in P. gingivalis-infected endothelial cells. However, the detailed pathological role of MIF has yet to be elucidated in this context. To explore the functional receptor(s) of MIF that underlie its participation in the pathogenesis of atherosclerosis, we investigated the expression of the chemokine receptors CD74 and CXCR4 in endothelial cells, both of which were shown to be involved in the adhesion of monocytes to endothelial cells pretreated with P. gingivalis. Furthermore, the formation of a MIF, CD74, and CXCR4 ligand-receptor complex was revealed by our immunofluorescence staining and coimmunoprecipitation results. By interacting with the CD74/CXCR4 receptor complex, MIF may act as a crucial regulator of monocyte-endothelial cell adhesion and promote the atherosclerotic plaque formation induced by P. gingivalis.
Collapse
Affiliation(s)
- Yun Wu
- Department of Periodontics, School of Stomatology, China Medical University, Heping District, Nanjing North Street No.117, Shenyang, 110002 China
| | - Wanyue Xu
- Department of Periodontics, School of Stomatology, China Medical University, Heping District, Nanjing North Street No.117, Shenyang, 110002 China
| | - Jingya Hou
- Department of Periodontics, School of Stomatology, China Medical University, Heping District, Nanjing North Street No.117, Shenyang, 110002 China
| | - Yanqing Liu
- Department of Periodontics, School of Stomatology, China Medical University, Heping District, Nanjing North Street No.117, Shenyang, 110002 China
| | - Rong Li
- Department of Periodontics, School of Stomatology, China Medical University, Heping District, Nanjing North Street No.117, Shenyang, 110002 China
| | - Jingbo Liu
- Department of Periodontics and Oral Biology, School of
Stomatology, China Medical University, Heping District, Nanjing North Street No.117, Shenyang, 110002 China
| | - Chen Li
- Department of Periodontics and Oral Biology, School of
Stomatology, China Medical University, Heping District, Nanjing North Street No.117, Shenyang, 110002 China
| | - Xiaolin Tang
- Department of Periodontics and Oral Biology, School of
Stomatology, China Medical University, Heping District, Nanjing North Street No.117, Shenyang, 110002 China
| | - Li Lin
- Department of Periodontics and Oral Biology, School of
Stomatology, China Medical University, Heping District, Nanjing North Street No.117, Shenyang, 110002 China
| | - Yaping Pan
- Department of Periodontics and Oral Biology, School of
Stomatology, China Medical University, Heping District, Nanjing North Street No.117, Shenyang, 110002 China
| | - Dongmei Zhang
- Department of Periodontics and Oral Biology, School of
Stomatology, China Medical University, Heping District, Nanjing North Street No.117, Shenyang, 110002 China
| |
Collapse
|
17
|
Forné C, Cambray S, Bermudez-Lopez M, Fernandez E, Bozic M, Valdivielso JM. Machine learning analysis of serum biomarkers for cardiovascular risk assessment in chronic kidney disease. Clin Kidney J 2019; 13:631-639. [PMID: 32905292 PMCID: PMC7467598 DOI: 10.1093/ckj/sfz094] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Accepted: 06/24/2019] [Indexed: 01/13/2023] Open
Abstract
Background Chronic kidney disease (CKD) patients show an increased burden of atherosclerosis and high risk of cardiovascular events (CVEs). There are several biomarkers described as being associated with CVEs, but their combined effectiveness in cardiovascular risk stratification in CKD has not been tested. The objective of this work is to analyse the combined ability of 19 biomarkers associated with atheromatous disease in predicting CVEs after 4 years of follow-up in a subcohort of the NEFRONA study in individuals with different stages of CKD without previous CVEs. Methods Nineteen putative biomarkers were quantified in 1366 patients (73 CVEs) and their ability to predict CVEs was ranked by random survival forest (RSF) analysis. The factors associated with CVEs were tested in Fine and Gray (FG) regression models, with non-cardiovascular death and kidney transplant as competing events. Results RSF analysis detected several biomarkers as relevant for predicting CVEs. Inclusion of those biomarkers in an FG model showed that high levels of osteopontin, osteoprotegerin, matrix metalloproteinase-9 and vascular endothelial growth factor increased the risk for CVEs, but only marginally improved the discrimination obtained with classical clinical parameters: concordance index 0.744 (95% confidence interval 0.609–0.878) versus 0.723 (0.592–0.854), respectively. However, in individuals with diabetes treated with antihypertensives and lipid-lowering drugs, the determination of these biomarkers could help to improve cardiovascular risk estimates. Conclusions We conclude that the determination of four biomarkers in the serum of CKD patients could improve cardiovascular risk prediction in high-risk individuals.
Collapse
Affiliation(s)
- Carles Forné
- Biostatistics Unit, Institute for Biomedical Research Dr. Pifarré Foundation, IRBLleida, Lleida, Spain.,Department of Basic Medical Sciences, University of Lleida, Lleida, Spain
| | - Serafi Cambray
- Vascular and Renal Translational Research Group, Institute for Biomedical Research Dr. Pifarré Foundation, IRBLleida and RedinRen RETIC, ISCIII, Lleida, Spain
| | - Marcelino Bermudez-Lopez
- Vascular and Renal Translational Research Group, Institute for Biomedical Research Dr. Pifarré Foundation, IRBLleida and RedinRen RETIC, ISCIII, Lleida, Spain
| | - Elvira Fernandez
- Vascular and Renal Translational Research Group, Institute for Biomedical Research Dr. Pifarré Foundation, IRBLleida and RedinRen RETIC, ISCIII, Lleida, Spain
| | - Milica Bozic
- Vascular and Renal Translational Research Group, Institute for Biomedical Research Dr. Pifarré Foundation, IRBLleida and RedinRen RETIC, ISCIII, Lleida, Spain
| | - Jose M Valdivielso
- Vascular and Renal Translational Research Group, Institute for Biomedical Research Dr. Pifarré Foundation, IRBLleida and RedinRen RETIC, ISCIII, Lleida, Spain
| | | |
Collapse
|
18
|
Garshick MS, Barrett T, Wechter T, Azarchi S, Scher J, Neimann A, Katz S, Fuentes-Duculan J, Cannizzaro MV, Jelic S, Fisher EA, Krueger JG, Berger JS. Inflammasome Signaling and Impaired Vascular Health in Psoriasis. Arterioscler Thromb Vasc Biol 2019; 39:787-798. [PMID: 30760013 PMCID: PMC6436998 DOI: 10.1161/atvbaha.118.312246] [Citation(s) in RCA: 60] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Objective- Psoriasis is an inflammatory skin disease which heightens the risk of cardiovascular disease. This study directly investigated vascular endothelial health and systemically altered pathways in psoriasis and matched controls. Approach and Results- Twenty patients (mean age, 40 years; 50% male) with active psoriasis and 10 age-, sex-matched controls were recruited. To investigate systemically alerted pathways, a deep sequencing omics approach was applied, including unbiased blood transcriptomic and targeted proteomic analysis. Vascular endothelial health was assessed by transcriptomic profiling of endothelial cells obtained from the brachial veins of recruited participants. Blood transcriptomic profiling identified inflammasome signaling as the highest differentially expressed canonical pathway ( Z score 1.6; P=1×10-7) including upregulation of CASP5 and interleukin ( IL) -1β. Proteomic panels revealed IL-6 as a top differentially expressed cytokine in psoriasis with pathway analysis highlighting IL-1β ( Z score 3.7; P=1.02×10-23) as an upstream activator of the observed upregulated proteins. Direct profiling of harvested brachial vein endothelial cells demonstrated inflammatory transcript (eg, IL-1β, CXCL10, VCAM-1, IL-8, CXCL1, Lymphotoxin beta, ICAM-1, COX-2, and CCL3) upregulation between psoriasis versus controls. A linear relationship was seen between differentially expressed endothelial inflammatory transcripts and psoriasis disease severity. IL-6 levels correlated with inflammatory endothelial cell transcripts and whole blood inflammasome-associated transcripts, including CASP5 and IL-1β. Conclusions- An unbiased sequencing approach demonstrated the inflammasome as the most differentially altered pathway in psoriasis versus controls. Inflammasome signaling correlated with psoriasis disease severity, circulating IL-6, and proinflammatory endothelial transcripts. These findings help better explain the heightened risk of cardiovascular disease in psoriasis. Clinical Trial Registration- URL: http://www.clinicaltrials.gov . Unique identifier: NCT03228017.
Collapse
Affiliation(s)
- Michael S. Garshick
- Center for the Prevention of Cardiovascular Disease, Department of Medicine, New York University School of Medicine;,Leon H. Charney Division of Cardiology, Department of Medicine, New York University School of Medicine
| | - Tessa Barrett
- Leon H. Charney Division of Cardiology, Department of Medicine, New York University School of Medicine
| | | | - Sarah Azarchi
- Ronald O. Perelman Department of Dermatology, New York University School of Medicine
| | - Jose Scher
- Psoriatic Arthritis Center, Division of Rheumatology, Department of Medicine, New York University School of Medicine
| | - Andrea Neimann
- Ronald O. Perelman Department of Dermatology, New York University School of Medicine
| | - Stuart Katz
- Leon H. Charney Division of Cardiology, Department of Medicine, New York University School of Medicine
| | | | | | - Sanja Jelic
- Division of Pulmonary, Allergy & Critical Care Medicine, Department of Medicine, Columbia University Medical Center
| | - Edward A. Fisher
- Center for the Prevention of Cardiovascular Disease, Department of Medicine, New York University School of Medicine;,Leon H. Charney Division of Cardiology, Department of Medicine, New York University School of Medicine
| | - James G. Krueger
- Laboratory for Investigative Dermatology, Rockefeller University
| | - Jeffrey S. Berger
- Center for the Prevention of Cardiovascular Disease, Department of Medicine, New York University School of Medicine;,Leon H. Charney Division of Cardiology, Department of Medicine, New York University School of Medicine;,Division of Vascular Surgery, Department of Surgery, New York University School of Medicine
| |
Collapse
|
19
|
Castro-Alves VC, Shiga TM, Nascimento JROD. Polysaccharides from chayote enhance lipid efflux and regulate NLRP3 inflammasome priming in macrophage-like THP-1 cells exposed to cholesterol crystals. Int J Biol Macromol 2019; 127:502-510. [PMID: 30658148 DOI: 10.1016/j.ijbiomac.2019.01.048] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2018] [Revised: 12/24/2018] [Accepted: 01/10/2019] [Indexed: 01/14/2023]
Abstract
The contribution of dietary fiber to decrease the risk of atherosclerosis may occur through other mechanisms besides the increased excretion of cholesterol. Although macrophages are crucial for lipid clearance, the excessive uptake of cholesterol crystals (CC) by these cells induce NLRP3 inflammasome and foam cell formation. Thus, we investigated whether the water-soluble DF from chayote (WSP) regulate CC-pretreated macrophage-like THP-1 cells. Linkage analysis indicated that WSP is composed mainly of pectic homogalacturonan and highly branched type I rhamnogalacturonan as well as hemicellulosic material including glucomannan, xyloglucan, and glucurono(arabino)xylan. WSP reduced interleukin (IL)-1β and chemokine release in CC-pretreated macrophages. Notably, WSP also reduced lipid accumulation in cells previously exposed to CC. Furthermore, WSP upregulated liver X receptor alpha expression, which may account for increased lipid efflux, and reduced matrix metallopeptidase 9 expression. WSP also reduced active caspase-1 protein levels, and downregulated NLRP3 and IL-1β gene expression in CC-pretreated cells, suggesting that this polysaccharide fraction regulates the priming signals required for NLRP3 inflammasome activation. Thus, WSP regulate lipid efflux and suppress inflammasome priming in macrophages, suggesting that the health benefits of this dietary fiber could go beyond its physical properties on the gastrointestinal tract.
Collapse
Affiliation(s)
- Victor Costa Castro-Alves
- Department of Food Science and Experimental Nutrition, School of Pharmaceutical Sciences, University of São Paulo, São Paulo, SP, Brazil; Food Research Center (FoRC), CEPID-FAPESP (Research, Innovation and Dissemination Centers, São Paulo Research Foundation), São Paulo, Brazil
| | - Tânia Misuzu Shiga
- Department of Food Science and Experimental Nutrition, School of Pharmaceutical Sciences, University of São Paulo, São Paulo, SP, Brazil; Food Research Center (FoRC), CEPID-FAPESP (Research, Innovation and Dissemination Centers, São Paulo Research Foundation), São Paulo, Brazil
| | - João Roberto Oliveira do Nascimento
- Department of Food Science and Experimental Nutrition, School of Pharmaceutical Sciences, University of São Paulo, São Paulo, SP, Brazil; Food Research Center (FoRC), CEPID-FAPESP (Research, Innovation and Dissemination Centers, São Paulo Research Foundation), São Paulo, Brazil; Food and Nutrition Research Center (NAPAN), University of São Paulo, São Paulo, Brazil.
| |
Collapse
|
20
|
Nuutinen S, Ailanen L, Savontaus E, Rinne P. Melanocortin overexpression limits diet-induced inflammation and atherosclerosis in LDLR -/- mice. J Endocrinol 2018; 236:111-123. [PMID: 29317531 DOI: 10.1530/joe-17-0636] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/03/2018] [Accepted: 01/09/2018] [Indexed: 12/26/2022]
Abstract
Atherosclerosis is a chronic inflammatory disease of the arteries. The disease is initiated by endothelial dysfunction that allows the transport of leukocytes and low-density lipoprotein into the vessel wall forming atherosclerotic plaques. The melanocortin system is an endogenous peptide system that regulates, for example, energy homeostasis and cardiovascular function. Melanocortin treatment with endogenous or synthetic melanocortin peptides reduces body weight, protects the endothelium and alleviates vascular inflammation, but the long-term effects of melanocortin system activation on atheroprogression remain largely unknown. In this study, we evaluated the effects of transgenic melanocortin overexpression in a mouse model of atherosclerosis. Low-density lipoprotein receptor-deficient mice overexpressing alpha- and gamma3-MSH (MSH-OE) and their wild-type littermates were fed either a regular chow or Western-style diet for 16 weeks. During this time, their metabolic parameters were monitored. The aortae were collected for functional analysis, and the plaques in the aortic root and arch were characterised by histological and immunohistochemical stainings. The aortic expression of inflammatory mediators was determined by quantitative PCR. We found that transgenic MSH-OE improved glucose tolerance and limited atherosclerotic plaque formation particularly in Western diet-fed mice. In terms of aortic vasoreactivity, MSH-OE blunted alpha1-adrenoceptor-mediated vasoconstriction and enhanced relaxation response to acetylcholine, indicating improved endothelial function. In addition, MSH-OE markedly attenuated Western diet-induced upregulation of proinflammatory cytokines (Ccl2, Ccl5 and Il6) that contribute to the pathogenesis of atherosclerosis. These results show that the activation of the melanocortin system improves glucose homeostasis and limits diet-induced vascular inflammation and atherosclerotic plaque formation.
Collapse
Affiliation(s)
- Salla Nuutinen
- Research Center for Integrative Physiology and Pharmacologyand Turku Center for Disease Modeling, Institute of Biomedicine, University of Turku, Turku, Finland
| | - Liisa Ailanen
- Research Center for Integrative Physiology and Pharmacologyand Turku Center for Disease Modeling, Institute of Biomedicine, University of Turku, Turku, Finland
| | - Eriika Savontaus
- Research Center for Integrative Physiology and Pharmacologyand Turku Center for Disease Modeling, Institute of Biomedicine, University of Turku, Turku, Finland
- Unit of Clinical PharmacologyTurku University Hospital, Turku, Finland
| | - Petteri Rinne
- Research Center for Integrative Physiology and Pharmacologyand Turku Center for Disease Modeling, Institute of Biomedicine, University of Turku, Turku, Finland
| |
Collapse
|
21
|
Abdulrazzaq Naji S, Jafarzadeh Kashi TS, Pourhajibagher M, Behroozibakhsh M, Masaeli R, Bahador A. Evaluation of Antimicrobial Properties of Conventional Poly(Methyl Methacrylate) Denture Base Resin Materials Containing Hydrothermally Synthesised Anatase TiO 2 Nanotubes against Cariogenic Bacteria and Candida albicans. IRANIAN JOURNAL OF PHARMACEUTICAL RESEARCH : IJPR 2018; 17:161-172. [PMID: 31011350 PMCID: PMC6447881] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
The purpose of this study was to investigate the antimicrobial properties of a conventional poly methyl methacrylate (PMMA) modified with hydrothermally synthesised titanium dioxide nanotubes (TNTs). Minimum inhibitory concentration (MIC), minimum bactericidal concentration (MBC), and minimum fungicidal concentrations (MFC) for planktonic cells of the TiO2 nanotubes solution against Lactobacillus acidophilus, Streptococcus mutans and Candida albicans were determined. The powder of conventional acrylic resin was modified using 2.5% and 5% by weight synthesised titanium dioxide (TiO2) nanotubes, and rectangular-shaped specimens (10 mm × 10 mm × 3 mm) were fabricated. The antimicrobial properties of ultraviolet (UV) and non-UV irradiated modified, and non-modified acrylic resins were evaluated using the estimation of planktonic cell count and biofilm formation of the three microorganisms mentioned above. The data were analysed by one-way analysis of variance (ANOVA), followed by a post-hoc Tukey's test at a significance level of 5%. MIC, for Streptococcus. mutans, Lactobacillus. acidophilus, and Candida. albicans, MBC for S. mutans and L. acidophilus and MFC for Candida. albicans were obtained more than 2100 µg/mL. The results of this study indicated a significant reduction in both planktonic cell count and biofilm formation of modified UV-activated acrylic specimens compared with the control group (p = 0.00). According to the results of the current study, it can be concluded that PMMA/TiO2 nanotube composite can be considered as a promising new material for antimicrobial approaches.
Collapse
Affiliation(s)
- Sahar Abdulrazzaq Naji
- Foundation of Technical Education, College of Health and Medical Technology, Baghdad, Iraq.,Department of Dental Biomaterials, School of Dentistry, International Campus, Tehran University of Medical Sciences (IC-TUMS), Tehran, Iran.
| | - Tahereh Sadat Jafarzadeh Kashi
- Department of Dental Biomaterials, School of Dentistry, Tehran University of Medical Sciences, Tehran, Iran. ,Dental Research Center, Dentistry Research Institute, Tehran University of Medical Sciences, Tehran, Iran.
| | - Maryam Pourhajibagher
- Dental Research Center, Dentistry Research Institute, Tehran University of Medical Sciences, Tehran, Iran.,Dental Implant Research Center, Dentistry Research Institute, Tehran University of Medical Sciences, Tehran, Iran.,Department of Microbiology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.
| | - Marjan Behroozibakhsh
- Department of Dental Biomaterials, School of Dentistry, Tehran University of Medical Sciences, Tehran, Iran. ,Research Center for Science and Technology in Medicine, Tehran University of Medical Sciences, Tehran, Iran.,Corresponding author: E-mail:
| | - Reza Masaeli
- Department of Dental Biomaterials, School of Dentistry, Tehran University of Medical Sciences, Tehran, Iran. ,Research Center for Science and Technology in Medicine, Tehran University of Medical Sciences, Tehran, Iran.
| | - Abbas Bahador
- Dental Research Center, Dentistry Research Institute, Tehran University of Medical Sciences, Tehran, Iran.,Department of Microbiology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
22
|
da Silva R, Fraga-Silva R, Steffens S, Fabre M, Bauer I, Caffa I, Magnone M, Sociali G, Quercioli A, Pelli G, Lenglet S, Galan K, Burger F, Calvo SV, Bertolotto M, Bruzzone S, Ballestrero A, Patrone F, Dallegri F, Santos R, Stergiopulos N, Mach F, Vuilleumier N, Montecucco F, Nencioni A. Nicotinamide phosphoribosyltransferase inhibition reduces intraplaque CXCL1 production and associated neutrophil infiltration in atherosclerotic mice. Thromb Haemost 2017; 111:308-22. [DOI: 10.1160/th13-07-0531] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2013] [Accepted: 09/30/2013] [Indexed: 11/05/2022]
Abstract
SummaryPharmacological treatments targeting CXC chemokines and the associated neutrophil activation and recruitment into atherosclerotic plaques hold promise for treating cardiovascular disorders. Therefore, we investigated whether FK866, a nicotinamide phosphoribosyltransferase (NAMPT) inhibitor with anti-inflammatory properties that we recently found to reduce neutrophil recruitment into the ischaemic myocardium, would exert beneficial effects in a mouse atherosclerosis model. Atherosclerotic plaque formation was induced by carotid cast implantation in ApoE-/- mice that were fed with a Western-type diet. FK866 or vehicle were administrated intraperitoneally from week 8 until week 11 of the diet. Treatment with FK866 reduced neutrophil infiltration and MMP-9 content and increased collagen levels in atherosclerotic plaques compared to vehicle. No effect on other histological parameters, including intraplaque lipids or macrophages, was observed. These findings were associated with a reduction in both systemic and intraplaque CXCL1 levels in FK866-treated mice. In vitro, FK866 did not affect MMP-9 release by neutrophils, but it strongly reduced CXCL1 production by endothelial cells which, in the in vivo model, were identified as a main CXCL1 source at the plaque level. CXCL1 synthesis inhibition by FK866 appears to reflect interference with nuclear factor-κB signalling as shown by reduced p65 nuclear levels in endothelial cells pre-treated with FK866. In conclusion, pharmacological inhibition of NAMPT activity mitigates inflammation in atherosclerotic plaques by reducing CXCL1-mediated activities on neutrophils. These results support further assessments of NAMPT inhibitors for the potential prevention of plaque vulnerability.
Collapse
|
23
|
Xie Z, Chan EC, Druey KM. R4 Regulator of G Protein Signaling (RGS) Proteins in Inflammation and Immunity. AAPS JOURNAL 2015; 18:294-304. [PMID: 26597290 DOI: 10.1208/s12248-015-9847-0] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/24/2015] [Accepted: 11/11/2015] [Indexed: 11/30/2022]
Abstract
G protein-coupled receptors (GPCRs) have important functions in both innate and adaptive immunity, with the capacity to bridge interactions between the two arms of the host responses to pathogens through direct recognition of secreted microbial products or the by-products of host cells damaged by pathogen exposure. In the mid-1990s, a large group of intracellular proteins was discovered, the regulator of G protein signaling (RGS) family, whose main, but not exclusive, function appears to be to constrain the intensity and duration of GPCR signaling. The R4/B subfamily--the focus of this review--includes RGS1-5, 8, 13, 16, 18, and 21, which are the smallest RGS proteins in size, with the exception of RGS3. Prominent roles in the trafficking of B and T lymphocytes and macrophages have been described for RGS1, RGS13, and RGS16, while RGS18 appears to control platelet and osteoclast functions. Additional G protein independent functions of RGS13 have been uncovered in gene expression in B lymphocytes and mast cell-mediated allergic reactions. In this review, we discuss potential physiological roles of this RGS protein subfamily, primarily in leukocytes having central roles in immune and inflammatory responses. We also discuss approaches to target RGS proteins therapeutically, which represents a virtually untapped strategy to combat exaggerated immune responses leading to inflammation.
Collapse
Affiliation(s)
- Zhihui Xie
- Molecular Signal Transduction Section, Laboratory of Allergic Diseases, NIAID/NIH, 50 South Drive Room 4154, Bethesda, Maryland, 20892, USA
| | - Eunice C Chan
- Molecular Signal Transduction Section, Laboratory of Allergic Diseases, NIAID/NIH, 50 South Drive Room 4154, Bethesda, Maryland, 20892, USA
| | - Kirk M Druey
- Molecular Signal Transduction Section, Laboratory of Allergic Diseases, NIAID/NIH, 50 South Drive Room 4154, Bethesda, Maryland, 20892, USA.
| |
Collapse
|
24
|
Tofik R, Swärd P, Ekelund U, Struglics A, Torffvit O, Rippe B, Bakoush O. Plasma pro-inflammatory cytokines, IgM-uria and cardiovascular events in patients with chest pain: A comparative study. Scandinavian Journal of Clinical and Laboratory Investigation 2015; 75:638-45. [PMID: 26174976 DOI: 10.3109/00365513.2015.1057218] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
BACKGROUND Risk stratification of patients presenting with acute chest pain is crucial for immediate and long-term management. Traditional predictors are suboptimal; therefore inflammatory biomarkers are studied for clinical assessment of patients at risk. Recently, we reported the association of IgM-uria with worse cardiovascular outcome in patients with acute chest pain. In this study, in the same cohort of patients with chest pain, we compared the value of IgM-uria to pro-inflammatory cytokines in predicting the occurrence of subsequent cardiovascular events. METHODS A total of 178 consecutive patients presenting with acute chest pain to the emergency department at the University Hospital of Lund, were recruited. Twenty-seven of 57 patients with acute coronary syndrome (ACS), and 18 of 118 patients with non-specific chest pain at baseline developed a subsequent major cardiovascular event during the 18 months follow-up. Urinary proteins (IgM-uria and Microalbuminuria) and plasma inflammatory markers (IL-6, Il-8, IL-10, IFN-γ and TNF-α) were measured at time of admission. RESULTS Using the receiver operating characteristic curves, the area under the curve for predicting cardiovascular events was 0.71 (95%CI 0.61-0.81) for IgM-uria, 0.61 (95%CI 0.51-0.71) for IL-6, 0.63 (95%CI 0.53-0.72) for IL-8, 0.65 (95%CI 0.56-0.74) for IL-10, and 0.64 (95% CI 0.54-0.74) for TNF-α. In multivariate Cox-regression analysis adjusted for age, microalbuminuria, IgM-uria, IL-10, TNF-α, troponin T, hsCRP and ACS at baseline; IgM-uria was the only biomarker that remained an independent predictor of outcome (HR = 4.2, 95%CI 2.2-7.8, p < 0.001). CONCLUSION In patients with chest pain with or without acute coronary syndrome, IgM-uria could better predict the occurrence of cardiovascular events than plasma pro-inflammatory cytokines.
Collapse
Affiliation(s)
- Rafid Tofik
- a Department of Emergency Medicine , Skåne University Hospital , Lund , Sweden.,b Department of Nephrology , Clinical Sciences Lund, Lund University , Lund , Sweden
| | - Per Swärd
- c Department of Orthopaedics , Clinical Sciences Lund, Lund University , Lund , Sweden
| | - Ulf Ekelund
- a Department of Emergency Medicine , Skåne University Hospital , Lund , Sweden
| | - André Struglics
- c Department of Orthopaedics , Clinical Sciences Lund, Lund University , Lund , Sweden
| | - Ole Torffvit
- b Department of Nephrology , Clinical Sciences Lund, Lund University , Lund , Sweden
| | - Bengt Rippe
- b Department of Nephrology , Clinical Sciences Lund, Lund University , Lund , Sweden
| | - Omran Bakoush
- b Department of Nephrology , Clinical Sciences Lund, Lund University , Lund , Sweden.,d Department of Internal Medicine , College of Medicine, UAE University , UAE
| |
Collapse
|
25
|
Wan W, Liu Q, Lionakis MS, Marino APMP, Anderson SA, Swamydas M, Murphy PM. Atypical chemokine receptor 1 deficiency reduces atherogenesis in ApoE-knockout mice. Cardiovasc Res 2015; 106:478-87. [PMID: 25858253 PMCID: PMC4447808 DOI: 10.1093/cvr/cvv124] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/15/2014] [Revised: 03/18/2015] [Accepted: 03/20/2015] [Indexed: 12/14/2022] Open
Abstract
AIMS Atypical chemokine receptor 1 (Ackr1; previously known as the Duffy antigen receptor for chemokines or Darc) is thought to regulate acute inflammatory responses in part by scavenging inflammatory CC and CXC chemokines; however, evidence for a role in chronic inflammation has been lacking. Here we investigated the role of Ackr1 in chronic inflammation, in particular in the setting of atherogenesis, using the apolipoprotein E-deficient (ApoE(-/-)) mouse model. METHODS AND RESULTS Ackr1(-/-)ApoE(-/-) and Ackr1(+/+)ApoE(-/-) littermates were obtained by crossing ApoE(-/-) mice and Ackr1(-/-) mice on a C57BL/6J background. Ackr1 (+/+)ApoE(-/-)mice fed a Western diet up-regulated Ackr1 expression in the aorta and had markedly increased atherosclerotic lesion size compared with Ackr1(-/-)ApoE(-/-) mice. This difference was observed in both the whole aorta and the aortic root in both early and late stages of the model. Ackr1 deficiency did not affect serum cholesterol levels or macrophage, collagen or smooth muscle cell content in atherosclerotic plaques, but significantly reduced the expression of Ccl2 and Cxcl1 in the whole aorta of ApoE(-/-) mice. In addition, Ackr1 deficiency resulted in a modest decrease in T cell subset frequency and inflammatory mononuclear phagocyte content in aorta and blood in the model. CONCLUSIONS Ackr1 deficiency appears to be protective in the ApoE knockout model of atherogenesis, but it is associated with only modest changes in cytokine and chemokine expression as well as T-cell subset frequency and inflammatory macrophage content.
Collapse
Affiliation(s)
- Wuzhou Wan
- Molecular Signaling Section, Laboratory of Molecular Immunology (LMI), National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD, USA
| | - Qian Liu
- Molecular Signaling Section, Laboratory of Molecular Immunology (LMI), National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD, USA
| | - Michail S Lionakis
- Fungal Pathogenesis Unit, Laboratory of Clinical Infectious Diseases, NIAID, NIH, Bethesda, MD, USA
| | - Ana Paula M P Marino
- Molecular Signaling Section, Laboratory of Molecular Immunology (LMI), National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD, USA
| | - Stasia A Anderson
- National Heart, Lung and Blood Institute (NHLBI) Animal MRI Core, NIH, Bethesda, MD, USA
| | - Muthulekha Swamydas
- Fungal Pathogenesis Unit, Laboratory of Clinical Infectious Diseases, NIAID, NIH, Bethesda, MD, USA
| | - Philip M Murphy
- Molecular Signaling Section, Laboratory of Molecular Immunology (LMI), National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD, USA
| |
Collapse
|
26
|
Abstract
Atherosclerosis is an inflammatory disease of the vessel wall characterized by activation of the innate immune system, with macrophages as the main players, as well as the adaptive immune system, characterized by a Th1-dominant immune response. Cytokines play a major role in the initiation and regulation of inflammation. In recent years, many studies have investigated the role of these molecules in experimental models of atherosclerosis. While some cytokines such as TNF or IFNγ clearly had atherogenic effects, others such as IL-10 were found to be atheroprotective. However, studies investigating the different cytokines in experimental atherosclerosis revealed that the cytokine system is complex with both disease stage-dependent and site-specific effects. In this review, we strive to provide an overview of the main cytokines involved in atherosclerosis and to shed light on their individual role during atherogenesis.
Collapse
Affiliation(s)
- Pascal J H Kusters
- Department of Medical Biochemistry, Academic Medical Center, Amsterdam, The Netherlands
| | - Esther Lutgens
- Department of Medical Biochemistry, Academic Medical Center, L01-146.1, University of Amsterdam, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands.
- Institute for Cardiovascular Prevention (IPEK), Ludwig Maximilians University (LMU), Munich, Germany.
| |
Collapse
|
27
|
Abstract
Atherosclerosis is one of the most common causes of death and disability in the United States today despite the availability of statins, which reduce hyperlipidemia, a risk factor that predisposes individuals to this disease. Epidemiology of human populations has overwhelmingly demonstrated an inverse correlation between the concentration of plasma high-density lipoprotein (HDL) cholesterol (HDL-C) and the likelihood of developing cardiovascular disease (CVD). Decades of observations and mechanistic studies suggest that one protective function of HDL is its central role in reverse cholesterol transport. In this pathway, the ATP-binding cassette transporter A1 releases intracellular cholesterol, which is packaged with apolipoprotein A-I (apoA-I) into nascent HDL particles and released from the plasma membrane. Further lipidation and maturation of HDL occur in plasma with the eventual uptake by the liver where cholesterol is removed. It is generally accepted that CVD risk can be reduced if plasma HDL-C levels are elevated. Several different pharmacological approaches have been tried; the most popular approach targets the movement of cholesteryl ester from HDL to triglyceride-rich particles by cholesteryl ester transfer protein. Inhibition of cholesteryl ester transfer protein increases plasma HDL-C concentration; however, beneficial effects have yet to be demonstrated, likely the result of off-target effects. These revelations have led to a reevaluation of how elevating HDL concentration could decrease risk. A recent, landmark study showed that the inherent cholesterol efflux capacity of an individual's plasma was a better predictor of CVD status than overall HDL-C concentration. Even more provocative are recent studies showing that apoA-I, the principle protein component of HDL modulates cellular inflammation and oxidation. The following will review all these potential routes explaining how HDL apoA-I can reduce the risk of CVD.
Collapse
|
28
|
Jara A, Benner CM, Sim D, Liu X, List EO, Householder LA, Berryman DE, Kopchick JJ. Elevated systolic blood pressure in male GH transgenic mice is age dependent. Endocrinology 2014; 155:975-86. [PMID: 24424040 PMCID: PMC3929738 DOI: 10.1210/en.2013-1899] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Acromegaly is associated with an increased incidence of cardiovascular disease. Transgenic mice expressing bovine GH (bGH) gene have previously been used to examine the effects of chronic GH stimulation on cardiovascular function. Results concerning systolic blood pressure (SBP) in bGH mice are conflicting. We hypothesized that these discrepancies may be the result of the various ages of the mice used in previous studies. In the current study, SBP was assessed monthly in male bGH mice from 3-12 months of age. Factors known to alter blood pressure were assessed during this time and included: levels of brain natriuretic peptide (BNP) and glucose homeostasis markers, and renal levels of angiotensin-converting enzyme 2 and endothelial nitric oxide synthase. Beginning at 6 months of age bGH had increased SBP compared with wild-type controls, which remained elevated through 12 months of age. Despite having increased blood pressure and cardiac BNP mRNA, bGH mice had decreased circulating levels of BNP. Additionally, bGH mice had an age-dependent decline in insulin levels. For example, they were hyperinsulinemic at 3 months, but by 11 months of age were hypoinsulinemic relative to wild-type controls. This decrease in insulin was accompanied by improved glucose tolerance at 11 months. Finally, both angiotensin-converting enzyme 2 and endothelial nitric oxide synthase expression were severely depressed in kidneys of 11-month-old bGH mice. These results indicate that elevated SBP in bGH mice is dependent on age, independent of insulin resistance, and related to alterations in both the natriuretic peptide and renin-angiotensin systems.
Collapse
Affiliation(s)
- Adam Jara
- Edison Biotechnology Institute (A.J., C.M.B., D.S., X.L., E.O.L., L.A.H., D.E.B., J.J.K.) Ohio University, Athens, Ohio 45701; Department of Biomedical Sciences (A.J., D.E.B., J.J.K.) and Department of Specialty Medicine (E.O.L.), Heritage College of Osteopathic Medicine, Athens, Ohio 45701; School of Applied Health Sciences and Wellness (C.M.B., X.L., D.E.B.) College of Health Sciences and Professions, Ohio University, Athens, Ohio 45701; and Department of Biological Sciences (A.J., J.J.K.) and Department of Social and Public Health (D.S.), Ohio University, Athens, Ohio 45701
| | | | | | | | | | | | | | | |
Collapse
|
29
|
Patel J, Channon KM, McNeill E. The downstream regulation of chemokine receptor signalling: implications for atherosclerosis. Mediators Inflamm 2013; 2013:459520. [PMID: 23690662 PMCID: PMC3649756 DOI: 10.1155/2013/459520] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2012] [Accepted: 03/12/2013] [Indexed: 12/13/2022] Open
Abstract
Heterotrimeric G-protein-coupled receptors (GPCRs) are key mediators of intracellular signalling, control numerous physiological processes, and are one of the largest class of proteins to be pharmacologically targeted. Chemokine-induced macrophage recruitment into the vascular wall is an early pathological event in the progression of atherosclerosis. Leukocyte activation and chemotaxis during cell recruitment are mediated by chemokine ligation of multiple GPCRs. Regulation of GPCR signalling is critical in limiting vascular inflammation and involves interaction with downstream proteins such as GPCR kinases (GRKs), arrestin proteins and regulator of G-protein signalling (RGS) proteins. These have emerged as new mediators of atherogenesis by functioning in internalisation, desensitisation, and signal termination of chemokine receptors. Targeting chemokine signalling through these proteins may provide new strategies to alter atherosclerotic plaque formation and plaque biology.
Collapse
Affiliation(s)
- Jyoti Patel
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, John Radcliffe Hospital, Oxford OX3 9DU, UK
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford OX3 7BN, UK
| | - Keith M. Channon
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, John Radcliffe Hospital, Oxford OX3 9DU, UK
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford OX3 7BN, UK
| | - Eileen McNeill
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, John Radcliffe Hospital, Oxford OX3 9DU, UK
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford OX3 7BN, UK
| |
Collapse
|