1
|
Zhang Y, Lu S, Qiu L, Qin M, Shan D, Xie L, Yi Y, Yu J. Integrative single-cell and bulk RNA-seq analyses identify CD4 + T-cell subpopulation infiltration and biomarkers of regulatory T cells involved in mediating the progression of atherosclerotic plaque. Front Immunol 2025; 15:1528475. [PMID: 39896809 PMCID: PMC11781991 DOI: 10.3389/fimmu.2024.1528475] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2024] [Accepted: 12/16/2024] [Indexed: 02/04/2025] Open
Abstract
Background Atherosclerosis (AS) is a chronic inflammatory disease with a significant contributor to mortality worldwide. Regulatory T cells (Tregs) are atheroprotective. However, the potential pathways and genes associated with atherosclerotic plaque progression in Tregs remain largely unknown. Therefore, this study aimed to identify critical target genes and pathways of Tregs associated with the progression of AS. Methods The gene expression data and single cell RNA-seq data of AS were downloaded from the Gene Expression Omnibus (GEO) database. Initially, we quantified CD4+ T cell proportions in non-plaque and plaque tissues using cell infiltration by estimation of RNA sequences (CIBERSORT) analysis, identifying pivotal transcription factors regulating the number of Tregs in atherosclerotic plaque. Subsequently, we identified significantly differential expressed genes of Tregs during the progression of atherosclerotic plaque and investigated the key pathways and transcription factors for these differentially expressed genes using gene ontology (GO) analysis and transcription factor enrichment analysis (TFEA), respectively. We also employed high dimensional weighted gene co-expression network analysis (hdWGCNA) and cell-cell communication analysis to elucidate the modules and cascade reaction of Tregs in the progression of AS. The key genes diagnostic potential was assessed via receiver operating characteristic (ROC) curve analysis. Finally, the target genes were validated in AS model using Ldlr-/- mice. Results We found that the proportion of Tregs significantly decreased, and Th2 cells showed a significant increase in atherosclerotic plaque compared to that in non-plaque arterial tissues. The five transcription factors (TEFC, IRF8, ZNF267, KLF2, and JUNB), identified as key targets associated with the function and the number of Tregs driving the progression of AS, primarily regulate immune response, ubiquitination, cytokine production, and T-cell differentiation pathways. ZNF267 may mainly involve in regulating ubiquitination, TGF-beta, and MAPK pathways of Tregs to regulate the function and the number of Tregs during the progress of AS. Interestingly, we found that IRF8 and ZNF267 as potential biomarkers were upregulated in circulating CD4+ T cells in patients with atherosclerotic coronary artery disease. Moreover, we also found that the changes of the function and the number of Tregs could modulate endothelial cell and smooth muscle cell functions to counteract AS through ligand-receptor pairs such as the MIF signaling pathway. Finally, we validated that two of the five transcription factors were also upregulated in mice atherosclerotic plaque through AS model using Ldlr-/- mice. Conclusion Our results indicate that the transcription factors TEFC, IRF8, ZNF267, KLF2, and JUNB in Tregs could be potential targets for the clinical management of AS.
Collapse
Affiliation(s)
- Yifeng Zhang
- Discipline of Chinese and Western Integrative Medicine, Jiangxi University of Chinese Medicine, Nanchang, China
- Centre for Translational Medicine, Jiangxi University of Chinese Medicine, Nanchang, China
| | - Shuxian Lu
- Discipline of Chinese and Western Integrative Medicine, Jiangxi University of Chinese Medicine, Nanchang, China
- Centre for Translational Medicine, Jiangxi University of Chinese Medicine, Nanchang, China
| | - Liang Qiu
- Discipline of Chinese and Western Integrative Medicine, Jiangxi University of Chinese Medicine, Nanchang, China
- Centre for Translational Medicine, Jiangxi University of Chinese Medicine, Nanchang, China
| | - Manman Qin
- Discipline of Chinese and Western Integrative Medicine, Jiangxi University of Chinese Medicine, Nanchang, China
- Centre for Translational Medicine, Jiangxi University of Chinese Medicine, Nanchang, China
| | - Dan Shan
- Department of Cardiovascular Sciences, Centre for Metabolic Disease Research, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, United States
| | - Lianhua Xie
- Discipline of Chinese and Western Integrative Medicine, Jiangxi University of Chinese Medicine, Nanchang, China
| | - Yao Yi
- Discipline of Chinese and Western Integrative Medicine, Jiangxi University of Chinese Medicine, Nanchang, China
- Institute of Obstetrics and Gynecology, Jiangxi University of Chinese Medicine, Nanchang, China
| | - Jun Yu
- Department of Cardiovascular Sciences, Centre for Metabolic Disease Research, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, United States
| |
Collapse
|
2
|
Chen J, Xu F, Mo X, Cheng Y, Wang L, Yang H, Li J, Zhang S, Zhang S, Li N, Cao Y. Exploratory Study of Differentially Expressed Genes of Peripheral Blood Monocytes in Patients with Carotid Atherosclerosis. Comb Chem High Throughput Screen 2024; 27:1344-1357. [PMID: 37608666 DOI: 10.2174/1386207326666230822122045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Revised: 06/15/2023] [Accepted: 07/13/2023] [Indexed: 08/24/2023]
Abstract
BACKGROUND The abundance of circulating monocytes is closely associated with the development of atherosclerosis in humans. OBJECTIVE This study aimed to further research into diagnostic biomarkers and targeted treatment of carotid atherosclerosis (CAS). METHODS We performed transcriptomics analysis through weighted gene co-expression network analysis (WGCNA) of monocytes from patients in public databases with and without CAS. Differentially expressed genes (DEGs) were screened by R package limma. Diagnostic molecules were derived by the least absolute shrinkage and selection operator (LASSO) and support vector machine recursive feature elimination (SVM-RFE) algorithms. NetworkAnalyst, miRWalk, and Star- Base databases assisted in the construction of diagnostic molecule regulatory networks. The Drug- Bank database predicted drugs targeting the diagnostic molecules. RT-PCR tested expression profiles. RESULTS From 14,369 hub genes and 61 DEGs, six differentially expressed monocyte-related hub genes were significantly associated with immune cells, immune responses, monocytes, and lipid metabolism. LASSO and SVM-RFE yielded five genes for CAS prediction. RT-PCR of these genes showed HMGB1 was upregulated, and CCL3, CCL3L1, CCL4, and DUSP1 were downregulated in CAS versus controls. Then, we constructed and visualized the regulatory networks of 9 transcription factors (TFs), which significantly related to 5 diagnostic molecules. About 11 miRNAs, 19 lncRNAs, and 39 edges centered on four diagnostic molecules (CCL3, CCL4, DUSP1, and HMGB1) were constructed and displayed. Eleven potential drugs were identified, including ibrutinib, CTI-01, roflumilast etc. Conclusion: A set of five biomarkers were identified for the diagnosis of CAS and for the study of potential therapeutic targets.
Collapse
Affiliation(s)
- Juhai Chen
- Guizhou Medical University, Guiyang, 550004, Guizhou Province, People's Republic of China
- Internal Medicine Department Three Ward, Guiyang Public Health Clinical Center, Guiyang, 550004, Guizhou Province, People's Republic of China
| | - Fengyan Xu
- Comprehensive Ward, Affiliated Hospital of Guizhou Medical University, Guiyang, 550004, Guizhou Province, People's Republic of China
| | - Xiangang Mo
- Comprehensive Ward, Affiliated Hospital of Guizhou Medical University, Guiyang, 550004, Guizhou Province, People's Republic of China
| | - Yiju Cheng
- The Department of Respiratory and Critical Medicine, Guiyang Public Health Clinical Center, Guiyang, 550004, Guizhou Province, People's Republic of China
| | - Lan Wang
- Comprehensive Ward, Affiliated Hospital of Guizhou Medical University, Guiyang, 550004, Guizhou Province, People's Republic of China
| | - Hui Yang
- Comprehensive Ward, Affiliated Hospital of Guizhou Medical University, Guiyang, 550004, Guizhou Province, People's Republic of China
| | - Jiajing Li
- Comprehensive Ward, Affiliated Hospital of Guizhou Medical University, Guiyang, 550004, Guizhou Province, People's Republic of China
| | - Shiyue Zhang
- Guizhou Medical University, Guiyang, 550004, Guizhou Province, People's Republic of China
| | - Shuping Zhang
- Guizhou Medical University, Guiyang, 550004, Guizhou Province, People's Republic of China
| | - Nannan Li
- Guizhou Medical University, Guiyang, 550004, Guizhou Province, People's Republic of China
| | - Yang Cao
- Guizhou Medical University, Guiyang, 550004, Guizhou Province, People's Republic of China
| |
Collapse
|
3
|
Huang R, Zhang L, Li X, Liu F, Cheng X, Ran H, Wang Z, Li Y, Feng Y, Liang L, Su W, Melgiri ND, Sun Y. Anti-CXCR2 antibody-coated nanoparticles with an erythrocyte-platelet hybrid membrane layer for atherosclerosis therapy. J Control Release 2023; 356:610-622. [PMID: 36898531 DOI: 10.1016/j.jconrel.2023.02.036] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 02/20/2023] [Accepted: 02/26/2023] [Indexed: 03/12/2023]
Abstract
Atherosclerosis is the leading cause of mortality globally. RBC-platelet hybrid membrane-coated nanoparticles ([RBC-P]NPs), which biologically mimic platelets in vivo, display evidence of anti-atherosclerotic activity. The efficacy of a targeted RBC-platelet hybrid membrane-coated nanoparticles ([RBC-P]NP)-based approach was investigated as a primary preventive measure against atherosclerosis. A ligand-receptor interactome analysis conducted with circulating platelets and monocytes derived from CAD patients and healthy controls identified CXCL8-CXCR2 as a key platelet ligand-monocyte receptor dyad in CAD patients. Based on this analysis, a novel anti-CXCR2 [RBC-P]NP that specifically binds to CXCR2 and blocks the interaction between CXCL8 and CXCR2 was engineered and characterized. Administering anti-CXCR2 [RBC-P]NPs to Western diet-fed Ldlr-/- mice led to diminished plaque size, necrosis, and intraplaque macrophage accumulation relative to control [RBC-P]NPs or vehicle. Importantly, anti-CXCR2 [RBC-P]NPs demonstrated no adverse bleeding/hemorrhagic effects. A series of in vitro experiments was conducted to characterize anti-CXCR2 [RBC-P]NP's mechanism of action in plaque macrophages. Mechanistically, anti-CXCR2 [RBC-P]NPs inhibited p38α (Mapk14)-mediated, pro-inflammatory M1 skewing and corrected efferocytosis in plaque macrophages. This targeted [RBC-P]NP-based approach, in which the cardioprotective effects of anti-CXCR2 [RBC-P]NP therapy overweighs its bleeding/hemorrhagic risks, could potentially be used to proactively manage atherosclerotic progression in at-risk populations.
Collapse
Affiliation(s)
- Rongzhong Huang
- Precision Medicine Center, The Second Affiliated Hospital of Chongqing Medical University, Chongqing Municipality Clinical Research Center for Geriatrics and Gerontology, Chongqing 400010, China
| | - Lujun Zhang
- Department of Cardiology, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Xingsheng Li
- Precision Medicine Center, The Second Affiliated Hospital of Chongqing Medical University, Chongqing Municipality Clinical Research Center for Geriatrics and Gerontology, Chongqing 400010, China
| | - Fan Liu
- Department of Ultrasound, The Second Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Ultrasound Molecular Imaging, Chongqing, China
| | - Xiaoxiao Cheng
- Department of Ultrasound, The Second Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Ultrasound Molecular Imaging, Chongqing, China
| | - Haitao Ran
- Department of Ultrasound, The Second Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Ultrasound Molecular Imaging, Chongqing, China
| | - Zhigang Wang
- Department of Ultrasound, The Second Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Ultrasound Molecular Imaging, Chongqing, China
| | - Yongyong Li
- Precision Medicine Center, The Second Affiliated Hospital of Chongqing Medical University, Chongqing Municipality Clinical Research Center for Geriatrics and Gerontology, Chongqing 400010, China
| | - Yuxing Feng
- Department of Rehabilitation and Pain Medicine, The Ninth People's Hospital of Chongqing, Chongqing, China
| | - Liwen Liang
- Department of Cardiology, The First People's Hospital of Yunnan Province, Kunming, China
| | - Wenhua Su
- Department of Cardiology, The First People's Hospital of Yunnan Province, Kunming, China
| | - N D Melgiri
- Impactys Foundation for Biomedical Research, San Diego, CA, USA
| | - Yang Sun
- Department of Ultrasound, The Second Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Ultrasound Molecular Imaging, Chongqing, China.
| |
Collapse
|
4
|
Qian Y, Zhang L, Sun Z, Zang G, Li Y, Wang Z, Li L. Biomarkers of Blood from Patients with Atherosclerosis Based on Bioinformatics Analysis. Evol Bioinform Online 2021; 17:11769343211046020. [PMID: 34594098 PMCID: PMC8477683 DOI: 10.1177/11769343211046020] [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] [Subscribe] [Scholar Register] [Received: 06/06/2021] [Accepted: 08/24/2021] [Indexed: 12/17/2022] Open
Abstract
Atherosclerosis is a multifaceted disease characterized by the formation and accumulation of plaques that attach to arteries and cause cardiovascular disease and vascular embolism. A range of diagnostic techniques, including selective coronary angiography, stress tests, computerized tomography, and nuclear scans, assess cardiovascular disease risk and treatment targets. However, there is currently no simple blood biochemical index or biological target for the diagnosis of atherosclerosis. Therefore, it is of interest to find a biochemical blood marker for atherosclerosis. Three datasets from the Gene Expression Omnibus (GEO) database were analyzed to obtain differentially expressed genes (DEG) and the results were integrated using the Robustrankaggreg algorithm. The genes considered more critical by the Robustrankaggreg algorithm were put into their own data set and the data set system with cell classification information for verification. Twenty-one possible genes were screened out. Interestingly, we found a good correlation between RPS4Y1, EIF1AY, and XIST. In addition, we know the general expression of these genes in different cell types and whole blood cells. In this study, we identified BTNL8 and BLNK as having good clinical significance. These results will contribute to the analysis of the underlying genes involved in the progression of atherosclerosis and provide insights for the discovery of new diagnostic and evaluation methods.
Collapse
Affiliation(s)
- Yongjiang Qian
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Lili Zhang
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Zhen Sun
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Guangyao Zang
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Yalan Li
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Zhongqun Wang
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Lihua Li
- Department of Pathology, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| |
Collapse
|
5
|
Lee CQE, Kerouanton B, Chothani S, Zhang S, Chen Y, Mantri CK, Hock DH, Lim R, Nadkarni R, Huynh VT, Lim D, Chew WL, Zhong FL, Stroud DA, Schafer S, Tergaonkar V, St John AL, Rackham OJL, Ho L. Coding and non-coding roles of MOCCI (C15ORF48) coordinate to regulate host inflammation and immunity. Nat Commun 2021; 12:2130. [PMID: 33837217 PMCID: PMC8035321 DOI: 10.1038/s41467-021-22397-5] [Citation(s) in RCA: 58] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Accepted: 03/10/2021] [Indexed: 02/01/2023] Open
Abstract
Mito-SEPs are small open reading frame-encoded peptides that localize to the mitochondria to regulate metabolism. Motivated by an intriguing negative association between mito-SEPs and inflammation, here we screen for mito-SEPs that modify inflammatory outcomes and report a mito-SEP named "Modulator of cytochrome C oxidase during Inflammation" (MOCCI) that is upregulated during inflammation and infection to promote host-protective resolution. MOCCI, a paralog of the NDUFA4 subunit of cytochrome C oxidase (Complex IV), replaces NDUFA4 in Complex IV during inflammation to lower mitochondrial membrane potential and reduce ROS production, leading to cyto-protection and dampened immune response. The MOCCI transcript also generates miR-147b, which targets the NDUFA4 mRNA with similar immune dampening effects as MOCCI, but simultaneously enhances RIG-I/MDA-5-mediated viral immunity. Our work uncovers a dual-component pleiotropic regulation of host inflammation and immunity by MOCCI (C15ORF48) for safeguarding the host during infection and inflammation.
Collapse
Affiliation(s)
- Cheryl Q. E. Lee
- grid.414735.00000 0004 0367 4692Institute of Medical Biology, A*STAR, Singapore, Singapore
| | - Baptiste Kerouanton
- grid.428397.30000 0004 0385 0924Duke-NUS Medical School, Program in Cardiovascular and Metabolic Disorders, Singapore, Singapore
| | - Sonia Chothani
- grid.428397.30000 0004 0385 0924Duke-NUS Medical School, Program in Cardiovascular and Metabolic Disorders, Singapore, Singapore
| | - Shan Zhang
- grid.428397.30000 0004 0385 0924Duke-NUS Medical School, Program in Cardiovascular and Metabolic Disorders, Singapore, Singapore
| | - Ying Chen
- grid.418812.60000 0004 0620 9243Institute of Molecular and Cell Biology, A*STAR, Singapore, Singapore
| | - Chinmay Kumar Mantri
- grid.428397.30000 0004 0385 0924Duke-NUS Medical School, Program in Emerging Infectious Diseases, Singapore, Singapore
| | - Daniella Helena Hock
- grid.1008.90000 0001 2179 088XDepartment of Biochemistry and Molecular Biology, The Bio21 Molecular Science & Biotechnology Institute, University of Melbourne, Melbourne, VIC Australia
| | - Radiance Lim
- grid.428397.30000 0004 0385 0924Duke-NUS Medical School, Program in Cardiovascular and Metabolic Disorders, Singapore, Singapore
| | - Rhea Nadkarni
- grid.428397.30000 0004 0385 0924Duke-NUS Medical School, Program in Cardiovascular and Metabolic Disorders, Singapore, Singapore
| | - Vinh Thang Huynh
- grid.418812.60000 0004 0620 9243Institute of Molecular and Cell Biology, A*STAR, Singapore, Singapore ,grid.59025.3b0000 0001 2224 0361Lee Kong Chian School of Medicine, Nanyang Technological University Singapore, Singapore, Singapore
| | - Daryl Lim
- grid.418377.e0000 0004 0620 715XGenome Institute Singapore, A*STAR, Singapore, Singapore
| | - Wei Leong Chew
- grid.418377.e0000 0004 0620 715XGenome Institute Singapore, A*STAR, Singapore, Singapore
| | - Franklin L. Zhong
- grid.59025.3b0000 0001 2224 0361Nanyang Technological University, Skin Diseases and Wound Repair Program, Singapore, Singapore ,grid.185448.40000 0004 0637 0221Skin Research Institute of Singapore, A*STAR, Singapore, Singapore
| | - David Arthur Stroud
- grid.1008.90000 0001 2179 088XDepartment of Biochemistry and Molecular Biology, The Bio21 Molecular Science & Biotechnology Institute, University of Melbourne, Melbourne, VIC Australia
| | - Sebastian Schafer
- grid.428397.30000 0004 0385 0924Duke-NUS Medical School, Program in Cardiovascular and Metabolic Disorders, Singapore, Singapore ,grid.419385.20000 0004 0620 9905National Heart Research Institute Singapore, National Heart Centre Singapore, Singapore, Singapore
| | - Vinay Tergaonkar
- grid.418812.60000 0004 0620 9243Institute of Molecular and Cell Biology, A*STAR, Singapore, Singapore ,grid.4280.e0000 0001 2180 6431Department of Pathology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Ashley L. St John
- grid.428397.30000 0004 0385 0924Duke-NUS Medical School, Program in Emerging Infectious Diseases, Singapore, Singapore ,grid.4280.e0000 0001 2180 6431Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore ,grid.189509.c0000000100241216Department of Pathology, Duke University Medical Center, Durham, NC USA
| | - Owen J. L. Rackham
- grid.428397.30000 0004 0385 0924Duke-NUS Medical School, Program in Cardiovascular and Metabolic Disorders, Singapore, Singapore
| | - Lena Ho
- grid.414735.00000 0004 0367 4692Institute of Medical Biology, A*STAR, Singapore, Singapore ,grid.428397.30000 0004 0385 0924Duke-NUS Medical School, Program in Cardiovascular and Metabolic Disorders, Singapore, Singapore ,grid.418812.60000 0004 0620 9243Institute of Molecular and Cell Biology, A*STAR, Singapore, Singapore
| |
Collapse
|
6
|
Zhang H, Prins J, Vreeken D, Florijn BW, de Bruin RG, van Hengel ORJ, van Essen MF, Duijs JMGJ, Van Esch H, van der Veer EP, van Zonneveld AJ, van Gils JM. Comprehensive analysis of neuronal guidance cue expression regulation during monocyte-to-macrophage differentiation reveals post-transcriptional regulation of semaphorin7A by the RNA-binding protein quaking. Innate Immun 2021; 27:118-132. [PMID: 33241976 PMCID: PMC7882812 DOI: 10.1177/1753425920966645] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2019] [Revised: 08/31/2020] [Accepted: 09/25/2020] [Indexed: 12/18/2022] Open
Abstract
In response to inflammatory cytokines and chemokines, monocytes differentiate into macrophages. Comprehensive analysis of gene expression regulation of neuronal guidance cue (NGC) ligands and receptors in the monocyte-to-macrophage differentiation process is not available yet. We performed transcriptome profiling in both human primary PBMCs/PBMC-derived macrophages and THP-1 cells/THP-1-macrophages using microarray or RNA sequencing methods. Pathway analysis showed that the axonal guidance pathway is significantly regulated upon monocyte differentiation. We confirmed NGC ligands and receptors which were consistently regulated, including SEMA4D, SEMA7A, NRP1, NRP2, PLXNA1 and PLXNA3. The involvement of RNA-binding protein quaking (QKI) in the regulation of NGC expression was investigated using monocytes and macrophages from a QKI haplo-insufficient patient and her healthy sibling. This revealed a positive correlation of SEMA7A expression with QKI expression. In silico analysis of 3'UTRs of NGCs proposed the competitive binding of QKI to proximal microRNA targeting sites as the mechanism of QKI-dependent regulation of SEMA7A. RNA immunoprecipitation confirmed an interaction of QKI with the 3'UTR of SEMA7A. Loss of SEMA7A resulted in monocyte differentiation towards a more anti-inflammatory macrophage. Taken together, the axonal guidance pathway is regulated during monocyte-to-macrophage differentiation, and the regulation is in line with the necessary functional adaption for the specialised role of macrophages.
Collapse
Affiliation(s)
- Huayu Zhang
- Einthoven Laboratory for Vascular and Regenerative Medicine, Department of Internal Medicine, Leiden University Medical Centre, The Netherlands
| | - Jurriën Prins
- Einthoven Laboratory for Vascular and Regenerative Medicine, Department of Internal Medicine, Leiden University Medical Centre, The Netherlands
| | - Dianne Vreeken
- Einthoven Laboratory for Vascular and Regenerative Medicine, Department of Internal Medicine, Leiden University Medical Centre, The Netherlands
| | - Barend W Florijn
- Einthoven Laboratory for Vascular and Regenerative Medicine, Department of Internal Medicine, Leiden University Medical Centre, The Netherlands
| | - Ruben G de Bruin
- Einthoven Laboratory for Vascular and Regenerative Medicine, Department of Internal Medicine, Leiden University Medical Centre, The Netherlands
| | - Oscar RJ van Hengel
- Einthoven Laboratory for Vascular and Regenerative Medicine, Department of Internal Medicine, Leiden University Medical Centre, The Netherlands
| | - Mieke F van Essen
- Einthoven Laboratory for Vascular and Regenerative Medicine, Department of Internal Medicine, Leiden University Medical Centre, The Netherlands
| | - Jacques MGJ Duijs
- Einthoven Laboratory for Vascular and Regenerative Medicine, Department of Internal Medicine, Leiden University Medical Centre, The Netherlands
| | - Hilde Van Esch
- Department of Human Genetics, University Hospitals Leuven, Belgium
| | - Eric P van der Veer
- Einthoven Laboratory for Vascular and Regenerative Medicine, Department of Internal Medicine, Leiden University Medical Centre, The Netherlands
| | - Anton Jan van Zonneveld
- Einthoven Laboratory for Vascular and Regenerative Medicine, Department of Internal Medicine, Leiden University Medical Centre, The Netherlands
| | - Janine M van Gils
- Einthoven Laboratory for Vascular and Regenerative Medicine, Department of Internal Medicine, Leiden University Medical Centre, The Netherlands
| |
Collapse
|
7
|
Zhang X, Lv X, Li X, Wang Y, Lin H, Zhang J, Peng C. Dysregulated circulating SOCS3 and haptoglobin expression associated with stable coronary artery disease and acute coronary syndrome: An integrated study based on bioinformatics analysis and case-control validation. Anatol J Cardiol 2020; 24:160-174. [PMID: 32870172 PMCID: PMC7585973 DOI: 10.14744/anatoljcardiol.2020.56346] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/17/2020] [Indexed: 02/05/2023] Open
Abstract
OBJECTIVE To extensively use blood transcriptome analysis to identify potential diagnostic and therapeutic targets for cardiovascular diseases. METHODS Two gene expression datasets (GSE59867 and GSE62646) were downloaded from GEO DataSets to identify altered blood transcriptomes in patients with ST-segment elevation myocardial infarction (STEMI) compared to stable coronary artery disease (CAD). Thereafter, several computational approaches were taken to determine functional roles and regulatory networks of differentially expressed genes (DEGs). Finally, the expression of dysregulated two hub genes-suppressor of cytokine signaling 3 (SOCS3) and haptoglobin (HP)-were validated in a case-control study. RESULTS A total of 119 DEGs were identified in the discovery phase, consisting of 71 downregulated genes and 48 upregulated genes; two hub modules consisting of two hub genes-SOCS3 and HP-were identified. In the validation phase, both SOCS3 and HP were significantly downregulated in the stable CAD and acute coronary syndrome (ACS) patients when compared with healthy controls. Meanwhile, HP was significantly upregulated in STEMI patients when compared with stable CAD patients (p=0.041). Logistic regression analysis indicated that: downregulated expression of HP correlated with increased risk of CAD [odds ratio (OR)=0.52, 95% confidence interval (CI)=0.31~0.87, p=0.013]; and downregulated expression of SOCS3 correlated with increased risk of ACS (OR=0.66, 95% CI=0.46~0.94, p=0.023) when age, gender, history of hyperlipidemia, diabetes and hypertension were included as covariates. CONCLUSION Future clarification of how SOCS3 and HP influence the pathogenesis of disease may pave the way for the development of novel diagnostic and therapeutic methods.
Collapse
Affiliation(s)
- Xunnan Zhang
- Postgraduate Training Basement of Jinzhou Medicical University, Taihe Hospital, Hubei University of Medicine; Hubei-P.R. China
| | - Xi Lv
- Postgraduate Training Basement of Jinzhou Medicical University, Taihe Hospital, Hubei University of Medicine; Hubei-P.R. China
| | - Xiandong Li
- Department of Laboratory Medicine, Taihe Hospital, Hubei University of Medicine; Hubei-P.R. China
| | - Yaping Wang
- Postgraduate Training Basement of Jinzhou Medicical University, Taihe Hospital, Hubei University of Medicine; Hubei-P.R. China
| | - Haoyu Lin
- Department of Breast and Thyroid Surgery, The First Affiliated Hospital of Shantou University Medical College; Guangdong-P.R. China
| | - Jicai Zhang
- Postgraduate Training Basement of Jinzhou Medicical University, Taihe Hospital, Hubei University of Medicine; Hubei-P.R. China
- Department of Laboratory Medicine, Taihe Hospital, Hubei University of Medicine; Hubei-P.R. China
| | - Chunyan Peng
- Department of Laboratory Medicine, Taihe Hospital, Hubei University of Medicine; Hubei-P.R. China
- Hubei Key Laboratory of Wudang Local Chinese Medicine Research, Hubei University of Medicine; Hubei-P.R. China
- Hubei Key Laboratory of Embryonic Stem Cell Research, Hubei University of Medicine, Hubei-P.R. China
- Address for correspondence: Chunyan Peng, MD, Department of Laboratory Medicine, Taihe Hospital, Hubei University of Medicine, Renming road 32# Shiyan, 442000, Hubei-P.R. China Phone: +86 13636 254788 E-mail:
| |
Collapse
|
8
|
Holvoet P, Klocke B, Vanhaverbeke M, Menten R, Sinnaeve P, Raitoharju E, Lehtimäki T, Oksala N, Zinser C, Janssens S, Sipido K, Lyytikainen LP, Cagnin S. RNA-sequencing reveals that STRN, ZNF484 and WNK1 add to the value of mitochondrial MT-COI and COX10 as markers of unstable coronary artery disease. PLoS One 2019; 14:e0225621. [PMID: 31821324 PMCID: PMC6903720 DOI: 10.1371/journal.pone.0225621] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2019] [Accepted: 11/09/2019] [Indexed: 12/23/2022] Open
Abstract
Markers in monocytes, precursors of macrophages, which are related to CAD, are largely unknown. Therefore, we aimed to identify genes in monocytes predictive of a new ischemic event in patients with CAD and/or discriminate between stable CAD and acute coronary syndrome. We included 66 patients with stable CAD, of which 24 developed a new ischemic event, and 19 patients with ACS. Circulating CD14+ monocytes were isolated with magnetic beads. RNA sequencing analysis in monocytes of patients with (n = 13) versus without (n = 11) ischemic event at follow-up and in patients with ACS (n = 12) was validated with qPCR (n = 85). MT-COI, STRN and COX10 predicted new ischemic events in CAD patients (power for separation at 1% error rate of 0.97, 0.90 and 0.77 respectively). Low MT-COI and high STRN were also related to shorter time between blood sampling and event. COX10 and ZNF484 together with MT-COI, STRN and WNK1 separated ACS completely from stable CAD patients. RNA expressions in monocytes of MT-COI, COX10, STRN, WNK1 and ZNF484 were independent of cholesterol lowering and antiplatelet treatment. They were independent of troponin T, a marker of myocardial injury. But, COX10 and ZNF484 in human plaques correlated to plaque markers of M1 macrophage polarization, reflecting vascular injury. Expression of MT-COI, COX10, STRN and WNK1, but not that of ZNF484, PBMCs paired with that in monocytes. The prospective study of relation of MT-COI, COX10, STRN, WNK1 and ZNF484 with unstable CAD is warranted.
Collapse
Affiliation(s)
- Paul Holvoet
- Department of Cardiovascular Sciences, KU Leuven, Leuven, Belgium
- * E-mail:
| | | | | | - Roxane Menten
- Department of Cardiovascular Sciences, KU Leuven, Leuven, Belgium
| | - Peter Sinnaeve
- Department of Clinical Cardiology, UZ Leuven, Leuven, Belgium
| | - Emma Raitoharju
- Department of Clinical Chemistry, Fimlab Laboratories, Tampere, Finland
- Finnish Cardiovascular Research Centre, Faculty of Medicine and Life Sciences University of Tampere, Tampere, Finland
| | - Terho Lehtimäki
- Department of Clinical Chemistry, Fimlab Laboratories, Tampere, Finland
- Finnish Cardiovascular Research Centre, Faculty of Medicine and Life Sciences University of Tampere, Tampere, Finland
| | - Niku Oksala
- Division of Vascular Surgery, Department of Surgery, Tampere University Hospital, Tampere, Finland
| | | | - Stefan Janssens
- Department of Clinical Cardiology, UZ Leuven, Leuven, Belgium
| | - Karin Sipido
- Department of Cardiovascular Sciences, KU Leuven, Leuven, Belgium
| | - Leo-Pekka Lyytikainen
- Department of Clinical Chemistry, Fimlab Laboratories, Tampere, Finland
- Finnish Cardiovascular Research Centre, Faculty of Medicine and Life Sciences University of Tampere, Tampere, Finland
| | - Stefano Cagnin
- Department of Biology, CRIBI Biotechnology Centre, Padova, Italy
- CIR-Myo Myology Centre, University of Padova, Padova, Italy
| |
Collapse
|
9
|
Carpentier S, Romagné F, Vivier E. A comprehensive approach to gene expression profiling in immune cells. Methods Enzymol 2019; 636:1-47. [PMID: 32178815 DOI: 10.1016/bs.mie.2019.07.005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/03/2022]
Abstract
With the advent of whole-transcriptome studies and the growing need for public repositories, it has become essential to combine multiple heterogeneous datasets for immune cells. In this chapter, we describe the implementation of a compendium of 10,833 genes for 975 samples, corresponding to 52 resting immune cell types. We begin by describing the datasets, and their selection, in particular. We then explain the methodology implemented to create a qualified compendium: the processing of each array (quality control, normalization and bias correction), integration (merging rules, global normalization and batch removal) and validation. Finally some examples of use will be detailed. The utility and limitations of the compendium are also discussed, as an introduction to the next version.
Collapse
Affiliation(s)
| | | | - Eric Vivier
- Innate Pharma Research Laboratories, Innate Pharma, Marseille, France; Aix-Marseille Univ, APHM, CNRS, INSERM, CIML, Hôpital de la Timone, Marseille-Immunopole, Marseille, France.
| |
Collapse
|
10
|
Swart M, Troeberg L. Effect of Polarization and Chronic Inflammation on Macrophage Expression of Heparan Sulfate Proteoglycans and Biosynthesis Enzymes. J Histochem Cytochem 2018; 67:9-27. [PMID: 30205019 DOI: 10.1369/0022155418798770] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Heparan sulfate (HS) proteoglycans on immune cells have the ability to bind to and regulate the bioactivity more than 400 bioactive protein ligands, including many chemokines, cytokines, and growth factors. This makes them important regulators of the phenotype and behavior of immune cells. Here we review how HS biosynthesis in macrophages is regulated during polarization and in chronic inflammatory diseases such as rheumatoid arthritis, atherosclerosis, asthma, chronic obstructive pulmonary disease and obesity, by analyzing published micro-array data and mechanistic studies in this area. We describe that macrophage expression of many HS biosynthesis and core proteins is strongly regulated by macrophage polarization, and that these expression patterns are recapitulated in chronic inflammation. Such changes in HS biosynthetic enzyme expression are likely to have a significant impact on the phenotype of macrophages in chronic inflammatory diseases by altering their interactions with chemokines, cytokines, and growth factors.
Collapse
Affiliation(s)
- Maarten Swart
- Kennedy Institute of Rheumatology, University of Oxford, Oxford, United Kingdom
| | - Linda Troeberg
- Kennedy Institute of Rheumatology, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
11
|
Guo J, Wang Q, Liu Y, Lu L, Hua Y, Hu R, Wang M, Li Z, Wang X, Wang BH, Fu Q, Chen A. Association of expression of ZNF606 gene from monocytes with the risk of coronary artery disease. Clin Biochem 2018; 60:44-51. [PMID: 30130524 DOI: 10.1016/j.clinbiochem.2018.08.005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2017] [Revised: 08/13/2018] [Accepted: 08/14/2018] [Indexed: 10/28/2022]
Abstract
AIM Messenger RNAs (mRNAs) play an important role in the pathogenesis of coronary artery disease (CAD). We evaluated the association of selected increase in mRNAs from monocytes with the risk of CAD. METHODS Chip data (GSE9820) retrieved from Gene Expression Omnibus (GEO) was re-analyzed, and the selected candidate genes, meeting specific conditions, were up-regulated and verified for specific biomarkers of CAD within a prospective cohort study that recruited 194 individuals and subdivided into two groups: group Non-CAD (GN), n = 68 and group CAD (GC), n = 126. The patients in GC were further categorized into three sub-units according to the extent of coronary stenosis shown during coronary angiography, coded as single-vessel stenosis (GC1, n = 53), 2-vessel stenosis (GC2, n = 50), or ≥ 3-vessel stenosis (GC3, n = 23). All candidate mRNAs expressions were analyzed from patients' monocytes with quantitative PCR (q-PCR). Receiver-operating characteristic (ROC) curves and the area under the ROC curves (AUCs) were used to evaluate the mRNAs' feasibility for CAD prediction. AUCs ≥0.8 were accounted as highly specific association with CAD. RESULTS GBA2, CSTF3, ZNF606 and MPP5 were selected as mRNAs candidates from chip data reanalysis. GBA2 (P = .002) and ZNF606 (P < .001) expressions were significantly increased in GC. ZNF606 showed significant increase after adjusting the risk factors with logistic regression analysis (OR = 3.804, 95% CI: 1.923, 7.798, P < .001), and its expression level was positively correlated with age (β = 0.04 × 10-3, P < .001). The AUCs (and 95% CI) of ZNF606 expression in GC2 and GC3 were ≥0.8. CONCLUSION These findings suggest that it is novel and specific for the association of ZNF606 gene expression from monocytes with the risk of CAD, especially in patients with multiple coronary artery stenosis.
Collapse
Affiliation(s)
- Jingbin Guo
- Department of Cardiology, Heart Center, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong 510282, China; Laboratory of Heart Center, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong 510282, China; Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular Disease, Guangzhou, Guangdong 510282, China; Sino-Japanese Cooperation Platform for Translational Research in Heart Failure, Guangzhou, Guangdong 510282, China
| | - Qiushi Wang
- Department of Cardiology, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, China
| | - Yangyang Liu
- The Huang-pu People's Hospital, Zhongshan, Guangdong 528403, China
| | - Lu Lu
- The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510407, China
| | - Yue Hua
- Monash Centre of Cardiovascular Research and Education in Therapeutics, Department of Epidemiology and Preventive Medicine, Monash University, Melbourne, Victoria 3004, Australia; School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Rong Hu
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Mingqing Wang
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Zhiliang Li
- Department of Cardiology, Heart Center, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong 510282, China; Laboratory of Heart Center, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong 510282, China; Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular Disease, Guangzhou, Guangdong 510282, China; Sino-Japanese Cooperation Platform for Translational Research in Heart Failure, Guangzhou, Guangdong 510282, China
| | - Xianbao Wang
- Department of Cardiology, Heart Center, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong 510282, China; Laboratory of Heart Center, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong 510282, China; Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular Disease, Guangzhou, Guangdong 510282, China; Sino-Japanese Cooperation Platform for Translational Research in Heart Failure, Guangzhou, Guangdong 510282, China
| | - Bing Hui Wang
- Monash Centre of Cardiovascular Research and Education in Therapeutics, Department of Epidemiology and Preventive Medicine, Monash University, Melbourne, Victoria 3004, Australia
| | - Qiang Fu
- Department of Cardiology, Heart Center, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong 510282, China; Laboratory of Heart Center, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong 510282, China; Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular Disease, Guangzhou, Guangdong 510282, China; Sino-Japanese Cooperation Platform for Translational Research in Heart Failure, Guangzhou, Guangdong 510282, China.
| | - Aihua Chen
- Department of Cardiology, Heart Center, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong 510282, China; Laboratory of Heart Center, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong 510282, China; Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular Disease, Guangzhou, Guangdong 510282, China; Sino-Japanese Cooperation Platform for Translational Research in Heart Failure, Guangzhou, Guangdong 510282, China.
| |
Collapse
|
12
|
Fleischer LM, Somaiya RD, Miller GM. Review and Meta-Analyses of TAAR1 Expression in the Immune System and Cancers. Front Pharmacol 2018; 9:683. [PMID: 29997511 PMCID: PMC6029583 DOI: 10.3389/fphar.2018.00683] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2017] [Accepted: 06/06/2018] [Indexed: 12/29/2022] Open
Abstract
Since its discovery in 2001, the major focus of TAAR1 research has been on its role in monoaminergic regulation, drug-induced reward and psychiatric conditions. More recently, TAAR1 expression and functionality in immune system regulation and immune cell activation has become a topic of emerging interest. Here, we review the immunologically-relevant TAAR1 literature and incorporate open-source expression and cancer survival data meta-analyses. We provide strong evidence for TAAR1 expression in the immune system and cancers revealed through NCBI GEO datamining and discuss its regulation in a spectrum of immune cell types as well as in numerous cancers. We discuss connections and logical directions for further study of TAAR1 in immunological function, and its potential role as a mediator or modulator of immune dysregulation, immunological effects of psychostimulant drugs of abuse, and cancer progression.
Collapse
Affiliation(s)
- Lisa M Fleischer
- Department of Pharmaceutical Sciences, Northeastern University, Boston, MA, United States
| | - Rachana D Somaiya
- Department of Pharmaceutical Sciences, Northeastern University, Boston, MA, United States
| | - Gregory M Miller
- Department of Pharmaceutical Sciences, Northeastern University, Boston, MA, United States.,Department of Chemical Engineering, Northeastern University, Boston, MA, United States.,Center for Drug Discovery, Northeastern University, Boston, MA, United States
| |
Collapse
|
13
|
Holvoet P, Vanhaverbeke M, Bloch K, Baatsen P, Sinnaeve P, Janssens S. Low MT-CO1 in Monocytes and Microvesicles Is Associated With Outcome in Patients With Coronary Artery Disease. J Am Heart Assoc 2016; 5:JAHA.116.004207. [PMID: 27919931 PMCID: PMC5210432 DOI: 10.1161/jaha.116.004207] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
BACKGROUND Cytochrome oxidase (COX) IV complex regulates energy production in mitochondria. Impaired COX gene expression is related to obesity and type 2 diabetes mellitus, but whether it is directly related to the incidence of cardiovascular events is unknown. We investigated whether COX gene expression in monocytes is predictive for cardiovascular events in coronary artery disease patients. To avoid monocyte isolation from fresh blood, we then aimed to validate our findings in monocyte-derived microvesicles isolated from plasma. METHODS AND RESULTS We enrolled 142 consecutive patients undergoing diagnostic coronary angiography between June 2010 and January 2011 and followed 67 patients with stable coronary artery disease prospectively for at least 3 years. Twenty-two patients experienced a new cardiovascular event (32.8%). Circulating CD14+ monocytes and microvesicles were isolated with magnetic beads, and COX mRNA levels were measured with quantitative polymerase chain reaction, after normalization with 5 validated house-keeping genes. Patients in the lowest tertile of mitochondrial cytochrome oxidase, subunit I (MT-COI) in monocytes at baseline had a higher risk for developing a new event after adjusting for age, sex, (ex)smoking, body mass index, blood pressure, diabetes mellitus, low-density lipoprotein- and high-density lipoprotein-cholesterol, triglycerides, high-sensitivity C-reactive protein, interleukin-6, and number of diseased vessels (harzard ratio [HR], 3.95; 95% CI, 1.63-9.57). Patients in the lowest tertile of MT-COI in monocyte-specific microvesicles had also a higher risk of developing a new event (adjusted HR, 5.00; 95% CI, 1.77-14). CONCLUSIONS In the current blinded study, low MT-COI in monocytes of coronary artery disease patients identifies a population at risk for new cardiovascular events. For the first time, we show that signatures in monocyte-specific microvesicles in plasma have similar predictive properties.
Collapse
Affiliation(s)
- Paul Holvoet
- Atherosclerosis and Metabolism Unit, Department of Cardiovascular Sciences, KU Leuven, Leuven, Belgium
| | | | | | | | - Peter Sinnaeve
- Department of Clinical Cardiology, UZ Leuven, Leuven, Belgium
| | - Stefan Janssens
- Department of Clinical Cardiology, UZ Leuven, Leuven, Belgium
| |
Collapse
|
14
|
Talukdar HA, Foroughi Asl H, Jain RK, Ermel R, Ruusalepp A, Franzén O, Kidd BA, Readhead B, Giannarelli C, Kovacic JC, Ivert T, Dudley JT, Civelek M, Lusis AJ, Schadt EE, Skogsberg J, Michoel T, Björkegren JLM. Cross-Tissue Regulatory Gene Networks in Coronary Artery Disease. Cell Syst 2016; 2:196-208. [PMID: 27135365 PMCID: PMC4855300 DOI: 10.1016/j.cels.2016.02.002] [Citation(s) in RCA: 103] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2015] [Revised: 12/02/2015] [Accepted: 01/30/2016] [Indexed: 01/23/2023]
Abstract
Inferring molecular networks can reveal how genetic perturbations interact with environmental factors to cause common complex diseases. We analyzed genetic and gene expression data from seven tissues relevant to coronary artery disease (CAD) and identified regulatory gene networks (RGNs) and their key drivers. By integrating data from genome-wide association studies, we identified 30 CAD-causal RGNs interconnected in vascular and metabolic tissues, and we validated them with corresponding data from the Hybrid Mouse Diversity Panel. As proof of concept, by targeting the key drivers AIP, DRAP1, POLR2I, and PQBP1 in a cross-species-validated, arterial-wall RGN involving RNA-processing genes, we re-identified this RGN in THP-1 foam cells and independent data from CAD macrophages and carotid lesions. This characterization of the molecular landscape in CAD will help better define the regulation of CAD candidate genes identified by genome-wide association studies and is a first step toward achieving the goals of precision medicine.
Collapse
Affiliation(s)
- Husain A Talukdar
- Cardiovascular Genomics Group, Division of Vascular Biology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, 171 77 Stockholm, Sweden
| | - Hassan Foroughi Asl
- Cardiovascular Genomics Group, Division of Vascular Biology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, 171 77 Stockholm, Sweden
| | - Rajeev K Jain
- Department of Physiology, Institute of Biomedicine and Translation Medicine, University of Tartu, 51014 Tartu, Estonia
| | - Raili Ermel
- Department of Physiology, Institute of Biomedicine and Translation Medicine, University of Tartu, 51014 Tartu, Estonia; Department of Cardiac Surgery, Tartu University Hospital, 51014 Tartu, Estonia
| | - Arno Ruusalepp
- Department of Physiology, Institute of Biomedicine and Translation Medicine, University of Tartu, 51014 Tartu, Estonia; Department of Cardiac Surgery, Tartu University Hospital, 51014 Tartu, Estonia; Clinical Gene Networks AB, 114 44 Stockholm, Sweden
| | - Oscar Franzén
- Clinical Gene Networks AB, 114 44 Stockholm, Sweden; Department of Genetics & Genomic Sciences, Institute of Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Brian A Kidd
- Department of Genetics & Genomic Sciences, Institute of Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Ben Readhead
- Department of Genetics & Genomic Sciences, Institute of Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Chiara Giannarelli
- Department of Genetics & Genomic Sciences, Institute of Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; The Zena and Michael A. Wiener Cardiovascular Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Jason C Kovacic
- The Zena and Michael A. Wiener Cardiovascular Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Torbjörn Ivert
- Department of Molecular Medicine and Surgery, Karolinska Institutet, 171 77 Stockholm, Sweden; Department of Thoracic Surgery, Karolinska University Hospital, 171 76 Stockholm, Sweden
| | - Joel T Dudley
- Department of Genetics & Genomic Sciences, Institute of Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Mete Civelek
- Departments of Medicine, Cardiology, Human Genetics, Microbiology, Immunology & Molecular Genetics, University of California, Los Angeles, Los Angeles, CA 90024, USA
| | - Aldons J Lusis
- Departments of Medicine, Cardiology, Human Genetics, Microbiology, Immunology & Molecular Genetics, University of California, Los Angeles, Los Angeles, CA 90024, USA
| | - Eric E Schadt
- Clinical Gene Networks AB, 114 44 Stockholm, Sweden; Department of Genetics & Genomic Sciences, Institute of Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Josefin Skogsberg
- Cardiovascular Genomics Group, Division of Vascular Biology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, 171 77 Stockholm, Sweden
| | - Tom Michoel
- Clinical Gene Networks AB, 114 44 Stockholm, Sweden; Division of Genetics and Genomics, The Roslin Institute, University of Edinburgh, Edinburgh EH25 9RG, UK
| | - Johan L M Björkegren
- Cardiovascular Genomics Group, Division of Vascular Biology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, 171 77 Stockholm, Sweden; Department of Physiology, Institute of Biomedicine and Translation Medicine, University of Tartu, 51014 Tartu, Estonia; Clinical Gene Networks AB, 114 44 Stockholm, Sweden; Department of Genetics & Genomic Sciences, Institute of Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Cardiovascular Institute, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.
| |
Collapse
|
15
|
Klaver EJ, van der Pouw Kraan TCTM, Laan LC, Kringel H, Cummings RD, Bouma G, Kraal G, van Die I. Trichuris suis soluble products induce Rab7b expression and limit TLR4 responses in human dendritic cells. Genes Immun 2015; 16:378-87. [PMID: 25996526 DOI: 10.1038/gene.2015.18] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2015] [Revised: 03/19/2015] [Accepted: 04/17/2015] [Indexed: 01/02/2023]
Abstract
Inflammatory immune disorders such as inflammatory bowel disease and multiple sclerosis are major health problems. Currently, the intestinal whipworm Trichuris suis is being explored in clinical trials to reduce inflammation in these diseases; however, the mechanisms by which the parasite affects the host immune system are not known. Here we determined the effects of T. suis soluble products (SPs) on Toll-like receptor-4 (TLR4)-stimulated human dendritic cells (DCs) using Illumina bead chip gene arrays. Pathway analysis of lipopolysaccharide-stimulated DCs with or without T. suis treatment showed that co-stimulation with T. suis SPs resulted in a downregulation of both the myeloid differentiation primary response gene 88-dependent and the TIR-domain-containing adaptor-inducing interferon-β-dependent signalling pathways triggered by TLR4. These data were verified using quantitative real-time PCR of several key genes within these pathways and/or defining their protein levels. In addition, T. suis SPs induce Rab7b, a negative regulator of TLR4 signalling that interferes with its trafficking, which coincided with a reduced surface expression of TLR4. These data indicate that the mechanism by which T. suis SPs reduce inflammatory responses is through suppression of both TLR4 signalling and surface expression on DCs.
Collapse
Affiliation(s)
- E J Klaver
- Department of Molecular Cell Biology and Immunology, VU University Medical Center, Amsterdam, The Netherlands
| | - T C T M van der Pouw Kraan
- Department of Molecular Cell Biology and Immunology, VU University Medical Center, Amsterdam, The Netherlands
| | - L C Laan
- Department of Molecular Cell Biology and Immunology, VU University Medical Center, Amsterdam, The Netherlands
| | - H Kringel
- Section for Parasitology, Health and Development, Department of Veterinary Disease Biology, University of Copenhagen, Copenhagen, Denmark
| | - R D Cummings
- Department of Biochemistry, Emory University School of Medicine, Atlanta, GA, USA
| | - G Bouma
- Department of Gastroenterology, VU University Medical Center, Amsterdam, The Netherlands
| | - G Kraal
- Department of Molecular Cell Biology and Immunology, VU University Medical Center, Amsterdam, The Netherlands
| | - I van Die
- Department of Molecular Cell Biology and Immunology, VU University Medical Center, Amsterdam, The Netherlands
| |
Collapse
|
16
|
Lee RH, Vazquez G. Reduced size and macrophage content of advanced atherosclerotic lesions in mice with bone marrow specific deficiency of alpha 7 nicotinic acetylcholine receptor. PLoS One 2015; 10:e0124584. [PMID: 25826262 PMCID: PMC4380454 DOI: 10.1371/journal.pone.0124584] [Citation(s) in RCA: 10] [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: 02/05/2015] [Accepted: 03/10/2015] [Indexed: 01/10/2023] Open
Abstract
In macrophages the α7 nicotinic acetylcholine receptor (α7nAChR) modulates production of inflammatory cytokines, cholesterol accumulation and lipoprotein uptake. Recently, our laboratory showed that selective stimulation of the α7nAChR protects macrophages from apoptosis, an effect that is absent in α7nAChR-deficient macrophages. All these observations are suggestive of a potential role of macrophage α7nAChR in atherosclerosis. Mouse models of the disease with bone marrow deletion of α7nAChR represent an attractive approach to address the in vivo relevance of these in vitro findings. However, recent studies that focused on the impact of hematopoietic deficiency of α7nAChR on early atherosclerotic lesions of low density lipoprotein receptor knockout (LDLRKO) mice, yielded controversial results. The question also remained whether macrophage α7nAChR modulates the characteristics of advanced lesions. Here we used LDLR knockout mice transplanted with bone marrow from wild-type or α7nAChR knockout animals to revisit the effect of hematopoietic deficiency of α7nAChR on early lesions and to examine, for the first time, its impact on advanced plaques. Aortic sinus atherosclerotic lesions were analyzed following 8 and 14 weeks on a high fat diet. Early lesions in mice with α7nAChR deficient bone marrow were not different from those in control animals. However, advanced lesions of mice with bone marrow deletion of α7nAChR exhibited reduction in size, macrophage content and cell proliferation. These studies are the first in examining the impact of hematopoietic deficiency of α7nAChR on the characteristics of advanced atherosclerotic lesions in a mouse model of the disease and provide novel evidence underscoring a potential pro-atherogenic role of macrophage α7nAChR.
Collapse
Affiliation(s)
- Robert H. Lee
- Department of Physiology and Pharmacology, Center for Diabetes and Endocrine Research, University of Toledo College of Medicine, Health Science Campus, 3000 Transverse Dr., Toledo, Ohio, 43614, United States of America
| | - Guillermo Vazquez
- Department of Physiology and Pharmacology, Center for Diabetes and Endocrine Research, University of Toledo College of Medicine, Health Science Campus, 3000 Transverse Dr., Toledo, Ohio, 43614, United States of America
- * E-mail:
| |
Collapse
|
17
|
The helminth Trichuris suis suppresses TLR4-induced inflammatory responses in human macrophages. Genes Immun 2014; 15:477-86. [PMID: 25008860 DOI: 10.1038/gene.2014.38] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2014] [Revised: 05/13/2014] [Accepted: 05/14/2014] [Indexed: 12/15/2022]
Abstract
Recent clinical trials in patients with inflammatory diseases like multiple sclerosis (MS) or inflammatory bowel disease (IBD) have shown the beneficial effects of probiotic helminth administration, although the underlying mechanism of action remains largely unknown. Potential cellular targets may include innate immune cells that propagate inflammation in these diseases, like pro-inflammatory macrophages. We here investigated the effects of the helminth Trichuris suis soluble products (SPs) on the phenotype and function of human inflammatory (granulocyte-macrophage colony-stimulating factor (GM-CSF)-differentiated) macrophages. Interestingly, we here show that T. suis SPs potently skew inflammatory macrophages into a more anti-inflammatory state in a Toll-like receptor 4 (TLR4)-dependent manner, and less effects are seen when stimulating macrophages with TLR2 or -3 ligands. Gene microarray analysis of GM-CSF-differentiated macrophages further revealed that many TLR4-induced inflammatory mediators, including interleukin (IL)-12B, CCL1 and CXCL9, are downregulated by T. suis SPs. In particular, we observed a strong reduction in the expression and function of P2RX7, a purinergic receptor involved in macrophage inflammation, leading to reduced IL-1β secretion. In conclusion, we show that T. suis SPs suppress a broad range of inflammatory pathways in GM-CSF-differentiated macrophages in a TLR4-dependent manner, thereby providing enhanced mechanistic insight into the therapeutic potential of this helminth for patients with inflammatory diseases.
Collapse
|
18
|
Systemic toll-like receptor and interleukin-18 pathway activation in patients with acute ST elevation myocardial infarction. J Mol Cell Cardiol 2014; 67:94-102. [PMID: 24389343 DOI: 10.1016/j.yjmcc.2013.12.021] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/09/2013] [Revised: 12/09/2013] [Accepted: 12/25/2013] [Indexed: 12/21/2022]
Abstract
Acute myocardial infarction (AMI) is accompanied by increased expression of Toll like receptors (TLR)-2 and TLR4 on circulating monocytes. In animal models, blocking TLR2/4 signaling reduces inflammatory cell influx and infarct size. The clinical consequences of TLR activation during AMI in humans are unknown, including its role in long-term cardiac functional outcome Therefore, we analyzed gene expression in whole blood samples from 28 patients with an acute ST elevation myocardial infarction (STEMI), enrolled in the EXenatide trial for AMI patients (EXAMI), both at admission and after 4-month follow-up, by whole genome expression profiling and real-time PCR. Cardiac function was determined by cardiac magnetic resonance (CMR) imaging at baseline and after 4-month follow-up. TLR pathway activation was shown by increased expression of TLR4 and its downstream genes, including IL-18R1, IL-18R2, IL-8, MMP9, HIF1A, and NFKBIA. In contrast, expression of the classical TLR-induced genes, TNF, was reduced. Bioinformatics analysis and in vitro experiments explained this noncanonical TLR response by identification of a pivotal role for HIF-1α. The extent of TLR activation and IL-18R1/2 expression in circulating cells preceded massive troponin-T release and correlated with the CMR-measured ischemic area (R=0.48, p=0.01). In conclusion, we identified a novel HIF-1-dependent noncanonical TLR activation pathway in circulating leukocytes leading to enhanced IL-18R expression which correlated with the magnitude of the ischemic area. This knowledge may contribute to our mechanistic understanding of the involvement of the innate immune system during STEMI and may yield diagnostic and prognostic value for patients with myocardial infarction.
Collapse
|
19
|
Hoefer IE, Sels JW, Jukema JW, Bergheanu S, Biessen E, McClellan E, Daemen M, Doevendans P, de Groot P, Hillaert M, Horsman S, Ilhan M, Kuiper J, Pijls N, Redekop K, van der Spek P, Stubbs A, van de Veer E, Waltenberger J, van Zonneveld AJ, Pasterkamp G. Circulating cells as predictors of secondary manifestations of cardiovascular disease: design of the CIRCULATING CELLS study. Clin Res Cardiol 2013; 102:847-56. [PMID: 23975238 DOI: 10.1007/s00392-013-0607-9] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/03/2013] [Accepted: 07/29/2013] [Indexed: 01/06/2023]
Abstract
Biomarkers for primary or secondary risk prediction of cardiovascular disease (CVD) are urgently needed to improve individual treatment and clinical trial design. The vast majority of biomarker discovery studies has concentrated on plasma/serum as an easily accessible source. Although numerous markers have been identified, their added predictive value on top of traditional risk factors has been limited, as the biological specimen does not specifically reflect expression profiles related with CVD progression and because the signal is often diluted by marker release from other organs. In contrast to serum markers, circulating cells serve as indicators of the actual disease state due to their active role in the pathogenesis of CVD and are responsible for the majority of secreted biomarkers. Therefore, the CIRCULATING CELLS study was initiated, focusing on the cellular effectors of atherosclerosis in the circulation. In total, 714 patients with coronary artery disease (CAD) symptoms were included. Blood cell fractions (monocytes, T-lymphocytes, platelets, granulocytes, PBMC) of all individual patients were isolated and stored for analysis. Concomitantly, extensive flow cytometric characterization of these populations was performed. From each patient, a detailed clinical profile together with extensive questionnaires about medical history and life style was obtained. Various high-throughput -omics approaches (protein, mRNA, miRNA) are currently being undertaken. Data will be integrated with advanced bioinformatics for discovery and validation of secondary risk markers for adverse events. Overall, the CIRCULATING CELLS study grants the interesting possibility that it will both identify novel biomarkers and provide useful insights into the pathophysiology of CAD in patients.
Collapse
Affiliation(s)
- Imo E Hoefer
- Laboratory of Experimental Cardiology, UMC Utrecht, Room G02.523, Heidelberglaan 100, 3584, Utrecht, CX, The Netherlands,
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Czepluch FS, Kuschicke H, Dellas C, Riggert J, Hasenfuss G, Schäfer K. Atheroprotective Krüppel-like factor 4 is downregulated in monocyte subsets of patients with coronary artery disease. Thromb Haemost 2013; 110:1080-2. [PMID: 23884216 DOI: 10.1160/th13-05-0367] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2013] [Accepted: 07/02/2013] [Indexed: 11/05/2022]
Affiliation(s)
- F S Czepluch
- Katrin Schäfer, MD, FESC, FAHA, Department of Cardiology and Pulmonary Medicine, University Medical Center Göttingen, Robert-Koch-Strasse 40, 37099 Göttingen, Germany, Tel.: +49 551 39 8921, Fax: +49 551 39 14131, E-mail:
| | | | | | | | | | | |
Collapse
|
21
|
Cho YS, Kim CH, Ha TS, Lee SJ, Ahn HY. Ginsenoside rg2 inhibits lipopolysaccharide-induced adhesion molecule expression in human umbilical vein endothelial cell. THE KOREAN JOURNAL OF PHYSIOLOGY & PHARMACOLOGY : OFFICIAL JOURNAL OF THE KOREAN PHYSIOLOGICAL SOCIETY AND THE KOREAN SOCIETY OF PHARMACOLOGY 2013; 17:133-7. [PMID: 23626475 PMCID: PMC3634090 DOI: 10.4196/kjpp.2013.17.2.133] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/02/2013] [Revised: 03/07/2013] [Accepted: 03/12/2013] [Indexed: 11/15/2022]
Abstract
Vascular cell adhesion molecule 1 (VCAM-1), intercellular adhesion molecule 1 (ICAM-1), P- and E-selectin play a pivotal role for initiation of atherosclerosis. Ginsenoside, a class of steroid glycosides, is abundant in Panax ginseng root, which has been used for prevention of illness in Korea. In this study, we investigated the mechanism(s) by which ginsenoside Rg2 may inhibit VCAM-1 and ICAM-1 expressions stimulated with lipopolysaccharide (LPS) in human umbilical vein endothelial cell (HUVEC). LPS increased VCAM-1 and ICAM-1 expression. Ginsenoside Rg2 prevented LPS-mediated increase of VCAM-1 and ICAM-1 expression. On the other hand, JSH, a nuclear factor kappa B (NF-κB) inhibitor, reduced both VCAM-1 and ICAM-1 expression stimulated with LPS. SB202190, inhibitor of p38 mitogen-activated protein kinase (p38 MAPK), and wortmannin, phosphatidylinositol 3-kinase (PI3-kinase) inhibitor, reduced LPS-mediated VCAM-1 but not ICAM-1 expression. PD98059, inhibitor of mitogen-activated protein kinase kinase/extracellular signal-regulated kinase (MEK/ERK) did not affect VCAM-1 and ICAM-1 expression stimulated with LPS. SP600125, inhibitor of c-Jun N-terminal kinase (JNK), reduced LPS-mediated ICAM-1 but not VCAM-1 expression. LPS reduced IkappaBα (IκBα) expression, in a time-dependent manner within 1 hr. Ginsenoside Rg2 prevented the decrease of IκBα expression stimulated with LPS. Moreover, ginsenoside Rg2 reduced LPS-mediated THP-1 monocyte adhesion to HUVEC, in a concentration-dependent manner. These data provide a novel mechanism where the ginsenoside Rg2 may provide direct vascular benefits with inhibition of leukocyte adhesion into vascular wall thereby providing protection against vascular inflammatory disease.
Collapse
Affiliation(s)
- Young-Suk Cho
- Department of Pharmacology, College of Medicine, Chungbuk National University, Cheongju 361-763, Korea
| | | | | | | | | |
Collapse
|
22
|
Manduteanu I, Simionescu M. Inflammation in atherosclerosis: a cause or a result of vascular disorders? J Cell Mol Med 2013; 16:1978-90. [PMID: 22348535 PMCID: PMC3822968 DOI: 10.1111/j.1582-4934.2012.01552.x] [Citation(s) in RCA: 95] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Sound data support the concept that in atherosclerosis, inflammation and dyslipidemia intersect each other and that irrespective of the initiator, both participate from the early stages to the ultimate fate of the atheromatous plaque. The two partakers manoeuvre a vicious circle in atheroma formation: dyslipidaemia triggers an inflammatory process and inflammation elicits dyslipidaemia. Independent of the initial cause, the atherosclerotic lesions occur focally, in particular arterial-susceptible sites, by a process that, although continuous, can be arbitrarily divided into a sequence of consecutive stages that lead from fatty streak to the fibro-lipid plaque and ultimately to plaque rupture and thrombosis. In the process, the initial event is a change in endothelial cells (EC) constitutive properties. Then, the molecular alarm signals send by dysfunctional EC are decoded by specific blood immune cells (monocytes, T lymphocytes, neutrophils, mast cells) and by the resident vascular cells, that respond by initiating a robust inflammatory process, in which the cells and the factors they secrete hasten the atheroma development. Direct and indirect crosstalk between the cells housed within the nascent plaque, complemented by the increase in risk factors of atherosclerosis lead to atheroma development and outcome. The initial inflammatory response can be regarded as a defense/protective reaction mechanism, but its further amplification, speeds up atherosclerosis. In this review, we provide an overview on the role of inflammation and dyslipidaemia and their intersection in atherogenesis. The data may add to the foundation of a novel attitude in the diagnosis and treatment of atherosclerosis.
Collapse
Affiliation(s)
- Ileana Manduteanu
- Institute of Cellular Biology and Pathology Nicolae Simionescu, Romanian Academy, Bucharest, Romania
| | | |
Collapse
|
23
|
Effects of omega-3 fatty acids on postprandial triglycerides and monocyte activation. Atherosclerosis 2012; 225:166-72. [DOI: 10.1016/j.atherosclerosis.2012.09.002] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/21/2012] [Revised: 09/04/2012] [Accepted: 09/04/2012] [Indexed: 11/24/2022]
|
24
|
Jie KE, van der Putten K, Wesseling S, Joles JA, Bergevoet MW, Pepers-de Kort F, Doevendans PA, Yasui Y, Liu Q, Verhaar MC, Gaillard CA, Braam B. Short-term erythropoietin treatment does not substantially modulate monocyte transcriptomes of patients with combined heart and renal failure. PLoS One 2012; 7:e41339. [PMID: 22957013 PMCID: PMC3434212 DOI: 10.1371/journal.pone.0041339] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2011] [Accepted: 06/25/2012] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND Combined heart and renal failure is associated with high cardiovascular morbidity and mortality. Anti-oxidant and anti-inflammatory, non-hematopoietic effects of erythropoietin (EPO) treatment have been proposed. Monocytes may act as biosensors of the systemic environment. We hypothesized that monocyte transcriptomes of patients with cardiorenal syndrome (CRS) reflect the pathophysiology of the CRS and respond to short-term EPO treatment at a recommended dose for treatment of renal anemia. METHODS Patients with CRS and anemia (n = 18) included in the EPOCARES trial were matched to healthy controls (n = 12). Patients were randomized to receive 50 IU/kg/week EPO or not. RNA from CD14(+)-monocytes was subjected to genome wide expression analysis (Illumina) at baseline and 18 days (3 EPO injections) after enrolment. Transcriptomes from patients were compared to healthy controls and effect of EPO treatment was evaluated within patients. RESULTS In CRS patients, expression of 471 genes, including inflammation and oxidative stress related genes was different from healthy controls. Cluster analysis did not separate patients from healthy controls. The 6 patients with the highest hsCRP levels had more differentially expressed genes than the 6 patients with the lowest hsCRP levels. Analysis of the variation in log(2) ratios of all individual 18 patients indicated that 4 of the 18 patients were different from the controls, whereas the other 14 were quite similar. After short-term EPO treatment, every patient clustered to his or her own baseline transcriptome. Two week EPO administration only marginally affected expression profiles on average, however, individual gene responses were variable. CONCLUSIONS In stable, treated CRS patients with mild anemia, monocyte transcriptomes were modestly altered, and indicated imprints of inflammation and oxidative stress. EPO treatment with a fixed dose has hematopoietic effects, had no appreciable beneficial actions on monocyte transcription profiles, however, could also not be associated with undesirable transcriptional responses.
Collapse
Affiliation(s)
- Kim E. Jie
- Department of Nephrology and Hypertension, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Karien van der Putten
- Department of Internal Medicine, Leiden University Medical Center, Leiden, The Netherlands
- Department of Internal Medicine, Meander Medical Center Amersfoort, Amersfoort, The Netherlands
| | - Sebastiaan Wesseling
- Department of Nephrology and Hypertension, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Jaap A. Joles
- Department of Nephrology and Hypertension, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Marloes W. Bergevoet
- Department of Nephrology and Hypertension, University Medical Center Utrecht, Utrecht, The Netherlands
| | | | - Pieter A. Doevendans
- Department of Cardiology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Yutaka Yasui
- Department of Public Health Sciences, School of Public Health, University of Alberta, Edmonton, Canada
| | - Qi Liu
- Department of Public Health Sciences, School of Public Health, University of Alberta, Edmonton, Canada
| | - Marianne C. Verhaar
- Department of Nephrology and Hypertension, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Carlo A. Gaillard
- Department of Internal Medicine, Meander Medical Center Amersfoort, Amersfoort, The Netherlands
- Department of Nephrology, Vrije Universiteit Medical Center, Amsterdam, The Netherlands
| | - Branko Braam
- Department of Medicine, Division of Nephrology and Immunology, University of Alberta, Edmonton, Canada
- Department of Physiology, University of Alberta, Edmonton, Canada
- * E-mail:
| |
Collapse
|
25
|
Sattler S, Reiche D, Sturtzel C, Karas I, Richter S, Kalb ML, Gregor W, Hofer E. The human C-type lectin-like receptor CLEC-1 is upregulated by TGF-β and primarily localized in the endoplasmic membrane compartment. Scand J Immunol 2012; 75:282-92. [PMID: 22117783 DOI: 10.1111/j.1365-3083.2011.02665.x] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
The orphan receptor CLEC-1 is part of a subfamily of C-type lectin-like receptors, which is encoded in the human natural killer gene complex and comprises several pattern recognition receptors important for innate immune functions. As information on human CLEC-1 is still very limited, we aimed to further characterize this receptor. Similar to another subfamily member, LOX-1, expression of CLEC-1 mRNA was detected in myeloid cells as well as in endothelial cells. CLEC-1 protein displayed N-linked glycosylation and formed dimers. However, in contrast to other members of the subfamily, expression levels were upregulated by transforming growth factor (TGF)-β, but not significantly affected by proinflammatory stimuli. It is intriguing that human CLEC-1 could only be detected intracellularly with a staining pattern resembling endoplasmic reticulum proteins. Neither TGF-β nor inflammatory stimuli could promote significant translocation to the cell surface. These findings are in accordance with a primarily intracellular localization and function of human CLEC-1.
Collapse
Affiliation(s)
- S Sattler
- Department of Vascular Biology and Thrombosis Research Immunology Dermatology, Division of Immunology, Allergy and Infectious Diseases, Medical University of Vienna, Vienna, Austria.
| | | | | | | | | | | | | | | |
Collapse
|
26
|
Golugula A, Lee G, Madabhushi A. Evaluating feature selection strategies for high dimensional, small sample size datasets. ANNUAL INTERNATIONAL CONFERENCE OF THE IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. ANNUAL INTERNATIONAL CONFERENCE 2012; 2011:949-52. [PMID: 22254468 DOI: 10.1109/iembs.2011.6090214] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
In this work, we analyze and evaluate different strategies for comparing Feature Selection (FS) schemes on High Dimensional (HD) biomedical datasets (e.g. gene and protein expression studies) with a small sample size (SSS). Additionally, we define a new feature, Robustness, specifically for comparing the ability of an FS scheme to be invariant to changes in its training data. While classifier accuracy has been the de facto method for evaluating FS schemes, on account of the curse of dimensionality problem, it might not always be the appropriate measure for HD/SSS datasets. SSS lends the dataset a higher probability of containing data that is not representative of the true distribution of the whole population. However, an ideal FS scheme must be robust enough to produce the same results each time there are changes to the training data. In this study, we employed the robustness performance measure in conjunction with classifier accuracy (measured via the K-Nearest Neighbor and Random Forest classifiers) to quantitatively compare five different FS schemes (T-test, F-test, Kolmogorov-Smirnov Test, Wilks Lambda Test and Wilcoxon Rand Sum Test) on 5 HD/SSS gene and protein expression datasets corresponding to ovarian cancer, lung cancer, bone lesions, celiac disease, and coronary heart disease. Of the five FS schemes compared, the Wilcoxon Rand Sum Test was found to outperform other FS schemes in terms of classification accuracy and robustness. Our results suggest that both classifier accuracy and robustness should be considered when deciding on the appropriate FS scheme for HD/SSS datasets.
Collapse
Affiliation(s)
- Abhishek Golugula
- Department of Electrical and Computer Engineering, Rutgers University, Piscataway, New Jersey 08854, USA
| | | | | |
Collapse
|
27
|
Kaminski WE, Beham AW, Kzhyshkowska J, Gratchev A, Puellmann K. On the horizon: flexible immune recognition outside lymphocytes. Immunobiology 2012; 218:418-26. [PMID: 22749215 DOI: 10.1016/j.imbio.2012.05.024] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2012] [Revised: 05/25/2012] [Accepted: 05/27/2012] [Indexed: 01/13/2023]
Abstract
Since decades there is consensus among immunologists that in jawless and jawed vertebrates flexible immune recognition is strictly confined to the lymphoid lineage. In jawed vertebrates the adaptive immune system is represented by two lineages of lymphocytes, B cells and T cells that express recombinatorial antigen receptors of enormous diversity known as immunoglobulins and the T cell receptor (TCR). The recent identification of recombined immune receptors that are structurally based on the TCR in subpopulations of neutrophils and eosinophils (referred to here as TCR-like immunoreceptors, "TCRL") provides unexpected evidence for the existence of flexible host defense mechanisms beyond the realm of lymphocytes. Consistent with this, subpopulations of monocytes and macrophages from humans and mice now have also been shown to constitutively express recombined TCR-like immunoreceptors. Available in vitro evidence suggests that the TCRL in macrophages may exert functions as facilitators of phagocytosis and self-recruitment. More importantly, our recent findings that the macrophage-TCRL is implicated in granuloma formation in tuberculosis and the neutrophil-TCRL is associated with autoimmune hemolytic anemia establish for the first time a link between myeloid recombinatorial immune receptors and clinical disease. The discovery of recombined TCR-like immune receptors in granulocytes and macrophages extends the principle of combinatorial immune recognition to phagocytic cells. Conceptually, this unifies the two hitherto disparate cardinal features of innate and adaptive immunity, phagocytic capacity and recombinatorial immune recognition on a common cellular platform. Moreover, it strongly suggests that flexible host defense in vertebrates may operate on a broader cellular basis than currently thought.
Collapse
Affiliation(s)
- Wolfgang E Kaminski
- Institute for Clinical Chemistry, University of Heidelberg Medical Faculty Mannheim, Mannheim, Germany.
| | | | | | | | | |
Collapse
|
28
|
Sivapalaratnam S, Basart H, Watkins NA, Maiwald S, Rendon A, Krishnan U, Sondermeijer BM, Creemers EE, Pinto-Sietsma SJ, Hovingh K, Ouwehand WH, Kastelein JJP, Goodall AH, Trip MD. Monocyte gene expression signature of patients with early onset coronary artery disease. PLoS One 2012; 7:e32166. [PMID: 22363809 PMCID: PMC3283726 DOI: 10.1371/journal.pone.0032166] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2011] [Accepted: 01/19/2012] [Indexed: 11/18/2022] Open
Abstract
The burden of cardiovascular disease (CVD) cannot be fully addressed by therapy targeting known pathophysiological pathways. Even with stringent control of all risk factors CVD events are only diminished by half. A number of additional pathways probably play a role in the development of CVD and might serve as novel therapeutic targets. Genome wide expression studies represent a powerful tool to identify such novel pathways. We compared the expression profiles in monocytes from twenty two young male patients with premature familial CAD with those from controls matched for age, sex and smoking status, without a family history of CVD. Since all patients were on statins and aspirin treatment, potentially affecting the expression of genes in monocytes, twelve controls were subsequently treated with simvastatin and aspirin for 6 and 2 weeks, respectively. By whole genome expression arrays six genes were identified to have differential expression in the monocytes of patients versus controls; ABCA1, ABCG1 and RGS1 were downregulated in patients, whereas ADRB2, FOLR3 and GSTM1 were upregulated. Differential expression of all genes, apart from GSTM1, was confirmed by qPCR. Aspirin and statins altered gene expression of ABCG1 and ADBR2. All finding were validated in a second group of twenty four patients and controls. Differential expression of ABCA1, RSG1 and ADBR2 was replicated. In conclusion, we identified these 3 genes to be expressed differently in CAD cases which might play a role in the pathogenesis of atherosclerotic vascular disease.
Collapse
|
29
|
Won HH, Kim SR, Bang OY, Lee SC, Huh W, Ko JW, Kim HG, McLeod HL, O'Connell TM, Kim JW, Lee SY. Differentially expressed genes in human peripheral blood as potential markers for statin response. J Mol Med (Berl) 2012; 90:201-211. [PMID: 21947165 DOI: 10.1007/s00109-011-0818-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2011] [Revised: 08/25/2011] [Accepted: 09/12/2011] [Indexed: 10/17/2022]
Abstract
There is a considerable inter-individual variation in response to statin therapy and one third of patients do not meet their treatment goals. We aimed to identify differentially expressed genes that might be involved in the effects of statin treatment and to suggest potential markers to guide statin therapy. Forty-six healthy Korean subjects received atorvastatin; their whole-genome expression profiles in peripheral blood were analyzed before and after atorvastatin administration in relation with changes in lipid profiles. The expression patterns of the differentially expressed genes were also compared with the data of familial hypercholesterolemia (FH) patients and controls. Pairwise comparison analyses revealed differentially expressed genes involved in diverse biological processes and molecular functions related with immune responses. Atorvastain mainly affected antigen binding, immune or inflammatory response including interleukin pathways. Similar expression patterns of the genes were observed in patients with FH and controls. The Charcol-Leyden crystal (CLC), CCR2, CX3CR1, LRRN3, FOS, LDLR, HLA-DRB1, ERMN, and TCN1 genes were significantly associated with cholesterol levels or statin response. Interestingly, the CLC gene, which was significantly altered by atorvastatin administration and differentially expressed between FH patients and controls, showed much bigger change in high-responsive group than in low-responsive group. We identified differentially expressed genes that might be involved in mechanisms underlying the known pleiotropic effects of atorvastatin, baseline cholesterol levels, and drug response. Our findings suggest CLC as a new candidate marker for statin response, and further validation is needed.
Collapse
Affiliation(s)
- Hong-Hee Won
- Samsung Biomedical Research Institute, Samsung Medical Center, Seoul 135-710, South Korea
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Yang H, Chen S, Tang Y, Dai Y. Interleukin-10 down-regulates oxLDL induced expression of scavenger receptor A and Bak-1 in macrophages derived from THP-1 cells. Arch Biochem Biophys 2011; 512:30-7. [DOI: 10.1016/j.abb.2011.05.017] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2011] [Revised: 05/01/2011] [Accepted: 05/23/2011] [Indexed: 01/09/2023]
|
31
|
Shalhoub J, Falck-Hansen MA, Davies AH, Monaco C. Innate immunity and monocyte-macrophage activation in atherosclerosis. JOURNAL OF INFLAMMATION-LONDON 2011; 8:9. [PMID: 21526997 PMCID: PMC3094203 DOI: 10.1186/1476-9255-8-9] [Citation(s) in RCA: 94] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/16/2010] [Accepted: 04/28/2011] [Indexed: 12/25/2022]
Abstract
Innate inflammation is a hallmark of both experimental and human atherosclerosis. The predominant innate immune cell in the atherosclerotic plaque is the monocyte-macrophage. The behaviour of this cell type within the plaque is heterogeneous and depends on the recruitment of diverse monocyte subsets. Furthermore, the plaque microenvironment offers polarisation and activation signals which impact on phenotype. Microenvironmental signals are sensed through pattern recognition receptors, including toll-like and NOD-like receptors - the latter of which are components of the inflammasome - thus dictating macrophage behaviour and outcome in atherosclerosis. Recently cholesterol crystals and modified lipoproteins have been recognised as able to directly engage these pattern recognition receptors. The convergent role of such pathways in terms of macrophage activation is discussed in this review.
Collapse
Affiliation(s)
- Joseph Shalhoub
- Cytokine Biology of Atherosclerosis, Kennedy Institute of Rheumatology, Faculty of Medicine, Imperial College London, UK.
| | | | | | | |
Collapse
|
32
|
Yang H, Chen SC. The effect of interleukin-10 on apoptosis in macrophages stimulated by oxLDL. Eur J Pharmacol 2011; 657:126-30. [PMID: 21296075 DOI: 10.1016/j.ejphar.2011.01.049] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2010] [Revised: 01/14/2011] [Accepted: 01/25/2011] [Indexed: 11/15/2022]
Abstract
Marked anti-atheromatous effects of the anti-inflammatory cytokine interleukin-10 (IL-10) were observed in several lipid-driven animal models of arteriosclerosis. We have previously demonstrated that IL-10 significantly inhibited lipid uptake in macrophages induced by oxLDL (Wang et al., 2008; Yang et al., 2008b). In this study, we investigated whether IL-10 affects the apoptosis related gene BCL2L11 and BMF expression in macrophages treated with oxLDL from THP-1 cells, which served as macrophage models. Cell apoptosis assays were performed by flow cytometry. Expression of the apoptosis related genes BCL2L11 and BMF mRNA was quantified by real-time RT-PCR (mRNA expression) and Western blotting (protein expression). IL-10 markedly blocked oxLDL induced cells undergoing early stage apoptosis. In the foam cell group, as compared with the macrophage group, the percentage of apoptosis increased by 100%. Here the expression of BCL2L11 was 45% (mRNA) and 41% (protein) elevated, while the expression of BMF was 54% (mRNA) and 44% (protein) elevated. When macrophages were co-stimulated by 100mg/l oxLDL and 20 μg/l IL-10 for 24h, compared with the foam cell group, the percentage of the apoptosis decreased by 21%, the expression of apoptosis related gene BMF was inhibited, the expression of mRNA and protein was both depressed by 23% and 20%, respectively, but the BCL2L11 expression was unchanged. These results may explain why decrement of early stage apoptosis cells was observed during co-stimulation and raise the possibility that IL-10 reduces foam cell undergoing apoptosis partly through down-regulating the expression of BMF, which demonstrates a critical role of IL-10 in anti-atherogenesis.
Collapse
Affiliation(s)
- Hong Yang
- Dept. of Biochemistry, Medical College of Tongji University, Shanghai 200092, China.
| | | |
Collapse
|
33
|
Abstract
Atherosclerosis is a chronic inflammatory disease occurring within the artery wall and is an underlying cause of cardiovascular complications, including myocardial infarction, stroke and peripheral vascular disease. Its pathogenesis involves many immune cell types with a well accepted role for monocyte/macrophages. Cholesterol-loaded macrophages are a characteristic feature of plaques and are major players in all stages of plaque development. As well as modulating lipid metabolism, macrophages secrete inflammatory cytokines, chemokines and reactive oxygen and nitrogen species that drive pathogenesis. They also produce proteases and tissue factor that contribute to plaque rupture and thrombosis. Macrophages are however heterogeneous cells and when appropriately activated, they phagocytose cytotoxic lipoproteins, clear apoptotic bodies, secrete anti-inflammatory cytokines and synthesize matrix repair proteins that stabilize vulnerable plaques. Pharmacological modulation of macrophage activity therefore represents a potential therapeutic strategy for atherosclerosis. The aim of this review is to provide an overview of the current understanding of the different macrophage subsets and their monocyte precursors, and, the implications of these subsets for atherosclerosis. This will present a foundation for highlighting novel opportunities to exploit the heterogeneity of macrophages as important diagnostic and therapeutic targets for atherosclerosis and its associated diseases.
Collapse
Affiliation(s)
- Heather M Wilson
- School of Medicine and Dentistry, University of Aberdeen, Institute of Medical Sciences, Foresterhill, Aberdeen, UK.
| |
Collapse
|
34
|
van der Pouw Kraan TCTM, Schirmer SH, Fledderus JO, Moerland PD, Baggen JM, Leyen TA, van der Laan AM, Piek JJ, van Royen N, Horrevoets AJG. Expression of a retinoic acid signature in circulating CD34 cells from coronary artery disease patients. BMC Genomics 2010; 11:388. [PMID: 20565948 PMCID: PMC2901320 DOI: 10.1186/1471-2164-11-388] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2009] [Accepted: 06/21/2010] [Indexed: 11/12/2022] Open
Abstract
Background Circulating CD34+ progenitor cells have the potential to differentiate into a variety of cells, including endothelial cells. Knowledge is still scarce about the transcriptional programs used by CD34+ cells from peripheral blood, and how these are affected in coronary artery disease (CAD) patients. Results We performed a whole genome transcriptome analysis of CD34+ cells, CD4+ T cells, CD14+ monocytes, and macrophages from 12 patients with CAD and 11 matched controls. CD34+ cells, compared to other mononuclear cells from the same individuals, showed high levels of KRAB box transcription factors, known to be involved in gene silencing. This correlated with high expression levels in CD34+ cells for the progenitor markers HOXA5 and HOXA9, which are known to control expression of KRAB factor genes. The comparison of expression profiles of CD34+ cells from CAD patients and controls revealed a less naïve phenotype in patients' CD34+ cells, with increased expression of genes from the Mitogen Activated Kinase network and a lowered expression of a panel of histone genes, reaching levels comparable to that in more differentiated circulating cells. Furthermore, we observed a reduced expression of several genes involved in CXCR4-signaling and migration to SDF1/CXCL12. Conclusions The altered gene expression profile of CD34+ cells in CAD patients was related to activation/differentiation by a retinoic acid-induced differentiation program. These results suggest that circulating CD34+ cells in CAD patients are programmed by retinoic acid, leading to a reduced capacity to migrate to ischemic tissues.
Collapse
Affiliation(s)
- Tineke C T M van der Pouw Kraan
- Department of Molecular Cell Biology and Immunology, VU University Medical Center, Van der Boechorststraat, 1081BT Amsterdam, The Netherlands.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Current world literature. Curr Opin Endocrinol Diabetes Obes 2010; 17:177-85. [PMID: 20190584 DOI: 10.1097/med.0b013e3283382286] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
|
36
|
Korybalska K, Pyda M, Grajek S, Łanocha M, Bręborowicz A, Witowski J. Serum profiles of monocyte chemoattractant protein-1 as a biomarker for patients recovering from myocardial infarction. Clin Res Cardiol 2010; 99:315-22. [DOI: 10.1007/s00392-010-0122-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/13/2009] [Accepted: 01/20/2010] [Indexed: 10/19/2022]
|
37
|
Abstract
PURPOSE OF REVIEW The varied behaviour of macrophages and foam cells during atherosclerosis and its clinical sequelae prompt the question whether all these activities can be the property of a single cell population. RECENT FINDINGS Subsets of monocytes with distinct patterns of surface markers and behaviours during inflammation have recently been characterized and shown to have complementary roles during progression of atherosclerosis. A variety of macrophage phenotypes derived from these monocyte subsets in response to mediators of innate and acquired immunity have also been found in plaques. Based on functional properties and genomic signatures, they may have different impacts on facets of plaque development, including fibrous cap and lipid core formation. SUMMARY Monocyte and macrophage phenotypic diversity is important in atherogenesis. More work is needed to define consistent marker sets for the different foam cell phenotypes in experimental animals and humans. Cell tracking studies are needed to establish their relationship with monocyte subtypes. In addition, genetic and pharmacological manipulation of phenotypes will be useful to define their functions and exploit the resulting therapeutic potential.
Collapse
Affiliation(s)
- Jason L Johnson
- Bristol Heart Institute, University of Bristol, Level 7, Bristol Royal Infirmary, Bristol BS2 8HW, UK
| | | |
Collapse
|
38
|
Sinnaeve PR, Donahue MP, Grass P, Seo D, Vonderscher J, Chibout SD, Kraus WE, Sketch M, Nelson C, Ginsburg GS, Goldschmidt-Clermont PJ, Granger CB. Gene expression patterns in peripheral blood correlate with the extent of coronary artery disease. PLoS One 2009; 4:e7037. [PMID: 19750006 PMCID: PMC2736586 DOI: 10.1371/journal.pone.0007037] [Citation(s) in RCA: 119] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2008] [Accepted: 08/09/2009] [Indexed: 11/19/2022] Open
Abstract
Systemic and local inflammation plays a prominent role in the pathogenesis of atherosclerotic coronary artery disease, but the relationship of whole blood gene expression changes with coronary disease remains unclear. We have investigated whether gene expression patterns in peripheral blood correlate with the severity of coronary disease and whether these patterns correlate with the extent of atherosclerosis in the vascular wall. Patients were selected according to their coronary artery disease index (CADi), a validated angiographical measure of the extent of coronary atherosclerosis that correlates with outcome. RNA was extracted from blood of 120 patients with at least a stenosis greater than 50% (CADi≥23) and from 121 controls without evidence of coronary stenosis (CADi = 0). 160 individual genes were found to correlate with CADi (rho>0.2, P<0.003). Prominent differential expression was observed especially in genes involved in cell growth, apoptosis and inflammation. Using these 160 genes, a partial least squares multivariate regression model resulted in a highly predictive model (r2 = 0.776, P<0.0001). The expression pattern of these 160 genes in aortic tissue also predicted the severity of atherosclerosis in human aortas, showing that peripheral blood gene expression associated with coronary atherosclerosis mirrors gene expression changes in atherosclerotic arteries. In conclusion, the simultaneous expression pattern of 160 genes in whole blood correlates with the severity of coronary artery disease and mirrors expression changes in the atherosclerotic vascular wall.
Collapse
Affiliation(s)
- Peter R Sinnaeve
- Duke University Medical Center and Duke Clinical Research Institute, Duke University, Durham, North Carolina, United States of America.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|