1
|
Li D, Pan L, Chen M, Zhang X, Jiang Z. TREM2 protects against LPS-induced murine acute lung injury through suppressing macrophage ferroptosis. Int Immunopharmacol 2025; 150:114247. [PMID: 39946766 DOI: 10.1016/j.intimp.2025.114247] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Revised: 02/05/2025] [Accepted: 02/05/2025] [Indexed: 03/03/2025]
Abstract
Triggering receptor expressed on myeloid cells-2 (TREM2) is a cell surface receptor, majorly expressed by granulocytes, monocytes and macrophages. We in this study showed that TREM2 was downregulated in the lipopolysaccharide (LPS)-treated macrophages and murine acute lung injury (ALI) through activation of p38 MAPK and STAT6 signaling. Over-expression of TREM2 reduced the expression of DNAX-activation protein 12 (DAP12), pro-inflammatory cytokines (TNF-α, IL-6, IL-1β), Malondialdehyde (MDA) and hemosiderin accumulation in LPS-treated macrophages. Knockdown of TREM2 expression elevated the expression of IL-6, reactive oxygen species (ROS), lactate dehydrogenases (LDH) and hemosiderin accumulation. Intratracheal adoptive transfer of TREM2-overexpressing macrophages effectively suppressed the lung inflammation and pro-inflammatory cytokine expression in murine ALI. While downregulation of TREM2 enhanced the lung inflammation in the lung tissues of murine ALI. Therefore, TREM2/DAP12 axis is involved in macrophage ferroptosis and attenuation of murine ALI. TREM2 would be a novel therapeutic target in murine ALI and patients with acute respiratory distress syndrome (ARDS).
Collapse
Affiliation(s)
- Dandan Li
- Department of Pulmonary and Critical Care Medicine, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou, China
| | - Linyue Pan
- Department of Pulmonary Medicine, Zhongshan Hospital Fudan University, Shanghai, China
| | - Mengjie Chen
- Department of Pulmonary Medicine, Zhongshan Hospital Fudan University, Shanghai, China
| | - Xiaoju Zhang
- Department of Pulmonary and Critical Care Medicine, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou, China
| | - Zhilong Jiang
- Department of Pulmonary Medicine, Zhongshan Hospital Fudan University, Shanghai, China.
| |
Collapse
|
2
|
Xie B, Pang S, Xie Y, Tan Q, Li S, Jili M, Huang Y, Zhao B, Yuan H, Mi J, Chen X, Ruan L, Chen H, Li X, Hu B, Huang J, Yang R, Li W. Urinary TYROBP and HCK as genetic biomarkers for non-invasive diagnosis and therapeutic targeting in IgA nephropathy. Front Genet 2024; 15:1516513. [PMID: 39777260 PMCID: PMC11703869 DOI: 10.3389/fgene.2024.1516513] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2024] [Accepted: 12/03/2024] [Indexed: 01/11/2025] Open
Abstract
Background IgA nephropathy (IgAN) is a leading cause of renal failure, but its pathogenesis remains unclear, complicating diagnosis and treatment. The invasive nature of renal biopsy highlights the need for non-invasive diagnostic biomarkers. Bulk RNA sequencing (RNA-seq) of urine offers a promising approach for identifying molecular changes relevant to IgAN. Methods We performed bulk RNA-seq on 53 urine samples from 11 untreated IgAN patients and 11 healthy controls, integrating these data with public renal RNA-seq, microarray, and scRNA-seq datasets. Machine learning was used to identify key differentially expressed genes, with protein expression validated by immunohistochemistry (IHC) and drug-target interactions explored via molecular docking. Results Urine RNA-seq analysis revealed differential expression profiles, from which TYROBP and HCK were identified as key biomarkers using machine learning. These biomarkers were validated in both a test cohort and an external validation cohort, demonstrating strong predictive accuracy. scRNA-seq confirmed their cell-specific expression patterns, correlating with renal function metrics such as GFR and serum creatinine. IHC further validated protein expression, and molecular docking suggested potential therapeutic interactions with IgAN treatments. Conclusion TYROBP and HCK are promising non-invasive urinary biomarkers for IgAN. Their predictive accuracy, validated through machine learning, along with IHC confirmation and molecular docking insights, supports their potential for both diagnostic and therapeutic applications in IgAN.
Collapse
Affiliation(s)
- Boji Xie
- Department of Nephrology, The Second Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Shuting Pang
- Department of Nephrology, The Second Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Yuli Xie
- Guangxi Key Laboratory for Genomic and Personalized Medicine, Center for Genomic and Personalized Medicine, Guangxi Collaborative Innovation Center for Genomic and Personalized Medicine, University Engineering Research Center of Digital Medicine and Healthcare, Guangxi Medical University, Nanning, Guangxi, China
- Department of Immunology, School of Basic Medical Sciences, Guangxi Medical University, Nanning, Guangxi, China
| | - Qiuyan Tan
- Department of Nephrology, The Second Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Shanshan Li
- Department of Nephrology, The Second Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Mujia Jili
- Guangxi Key Laboratory for Genomic and Personalized Medicine, Center for Genomic and Personalized Medicine, Guangxi Collaborative Innovation Center for Genomic and Personalized Medicine, University Engineering Research Center of Digital Medicine and Healthcare, Guangxi Medical University, Nanning, Guangxi, China
- Department of Immunology, School of Basic Medical Sciences, Guangxi Medical University, Nanning, Guangxi, China
| | - Yian Huang
- Guangxi Key Laboratory for Genomic and Personalized Medicine, Center for Genomic and Personalized Medicine, Guangxi Collaborative Innovation Center for Genomic and Personalized Medicine, University Engineering Research Center of Digital Medicine and Healthcare, Guangxi Medical University, Nanning, Guangxi, China
- Department of Immunology, School of Basic Medical Sciences, Guangxi Medical University, Nanning, Guangxi, China
| | - Binran Zhao
- Department of Nephrology, The Second Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Hao Yuan
- Guangxi Key Laboratory for Genomic and Personalized Medicine, Center for Genomic and Personalized Medicine, Guangxi Collaborative Innovation Center for Genomic and Personalized Medicine, University Engineering Research Center of Digital Medicine and Healthcare, Guangxi Medical University, Nanning, Guangxi, China
- Department of Immunology, School of Basic Medical Sciences, Guangxi Medical University, Nanning, Guangxi, China
| | - Junhao Mi
- Guangxi Key Laboratory for Genomic and Personalized Medicine, Center for Genomic and Personalized Medicine, Guangxi Collaborative Innovation Center for Genomic and Personalized Medicine, University Engineering Research Center of Digital Medicine and Healthcare, Guangxi Medical University, Nanning, Guangxi, China
- Department of Immunology, School of Basic Medical Sciences, Guangxi Medical University, Nanning, Guangxi, China
- Medical Laboratory Department, Liuzhou Maternity and Child Healthcare Hospital, Liuzhou, China
| | - Xuesong Chen
- Department of Nephrology, The Second Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Liangping Ruan
- Department of Nephrology, The Second Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Hong Chen
- Department of Nephrology, The Second Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Xiaolai Li
- Department of Nephrology, The Second Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Boning Hu
- Department of Nephrology, The Second Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Jing Huang
- Department of Nephrology, The Second Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Rirong Yang
- Guangxi Key Laboratory for Genomic and Personalized Medicine, Center for Genomic and Personalized Medicine, Guangxi Collaborative Innovation Center for Genomic and Personalized Medicine, University Engineering Research Center of Digital Medicine and Healthcare, Guangxi Medical University, Nanning, Guangxi, China
- Department of Immunology, School of Basic Medical Sciences, Guangxi Medical University, Nanning, Guangxi, China
| | - Wei Li
- Department of Nephrology, The Second Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| |
Collapse
|
3
|
Luo H, Pan C, Wang L, Zheng L, Cao S, Hu X, Hu T, Zhao N, Shang Q, Wang J. Low TYROBP expression predicts poor prognosis in multiple myeloma. Cancer Cell Int 2024; 24:117. [PMID: 38549127 PMCID: PMC10979612 DOI: 10.1186/s12935-024-03304-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2023] [Accepted: 03/14/2024] [Indexed: 04/01/2024] Open
Abstract
BACKGROUND Multiple myeloma (MM) is the second most common refractory hematologic cancer. Searching for new targets and prognostic markers for MM is significant. METHODS GSE39754, GSE6477 and GSE24080 were downloaded from the Gene Expression Omnibus (GEO) database. Differentially expressed genes (DEGs) in MM versus healthy people from GSE39754 and GSE6477 were screened using limma package, and MM-related module genes were chosen with the use of Weighted gene co-expression network analysis (WGCNA), and the two were intersected using ggVennDiagram for obtaining MM-related DEGs. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) analyses were carried out. Then, protein-protein interactions (PPI) analysis in String database was used to obtain hub genes, while prognosis was analyzed by survival package in GSE24080. Receiver operating characteristic (ROC) curve was adopted for evaluating diagnostic value of hub genes. Besides, univariable/multivariable Cox regression were employed to screen independent prognostic biomarkers. Gene set enrichment analysis (GSEA) was used to find possible mechanism. Finally, western-blotting and reverse transcription-polymerase chain reaction (RT-PCR) verify TYROBP expression within MM and healthy people. We performed cell adhesion and transwell assays for investigating TYROBP function in MM cell adhesion and migration. RESULTS Through differential analyses, 92 MM-related DEGs were obtained. 10 hub genes were identified by PPI and CytoHubba. Their diagnostic and prognostic significance was analyzed. Down-regulation of genes like TYROBP, ELANE, MNDA, and MPO related to dismal MM prognosis. Upon univariable/multivariable Cox regression, TYROBP independently predicted MM prognosis. GSEA pathway was enriched, indicating that TYROBP expression affected MM development via cell adhesion molecular pathway. Upon Western-blotting and RT-PCR assays, TYROBP expression among MM patients decreased relative to healthy donors. Cell adhesion and transwell migration assays revealed increased MM cell adhesion and decreased migration upon TYROBP up-regulation. CONCLUSION In summary, TYROBP is a potential prognostic marker for MM.
Collapse
Affiliation(s)
- Hong Luo
- Department of Hematology, Affiliated Hospital of Guizhou Medical University, Guiyang, 550004, China
- Department of Clinical Medical School, Guizhou Medical University, Guiyang, 550004, China
| | - Chengyun Pan
- Department of Hematology, Affiliated Hospital of Guizhou Medical University, Guiyang, 550004, China
| | - Li Wang
- Department of Hematology, Affiliated Hospital of Guizhou Medical University, Guiyang, 550004, China
- Department of Clinical Medical School, Guizhou Medical University, Guiyang, 550004, China
| | - Lin Zheng
- Department of Hematology, Affiliated Hospital of Guizhou Medical University, Guiyang, 550004, China
- Department of Clinical Medical School, Guizhou Medical University, Guiyang, 550004, China
| | - Shuyun Cao
- Department of Hematology, Affiliated Hospital of Guizhou Medical University, Guiyang, 550004, China
- Department of Clinical Medical School, Guizhou Medical University, Guiyang, 550004, China
| | - Xiuying Hu
- Department of Hematology, Affiliated Hospital of Guizhou Medical University, Guiyang, 550004, China
| | - Tianzhen Hu
- Department of Hematology, Affiliated Hospital of Guizhou Medical University, Guiyang, 550004, China
| | - Naiqin Zhao
- Department of Hematology, Affiliated Hospital of Guizhou Medical University, Guiyang, 550004, China
| | - Qin Shang
- Department of Hematology, Affiliated Hospital of Guizhou Medical University, Guiyang, 550004, China
| | - Jishi Wang
- Department of Hematology, Affiliated Hospital of Guizhou Medical University, Guiyang, 550004, China.
- Department of Clinical Medical School, Guizhou Medical University, Guiyang, 550004, China.
- National Clinical Research Center for Hematologic Diseases, The First Affiliated Hospital of Soochow University, Jiangsu, 215006, China.
| |
Collapse
|
4
|
Abstract
Triggering receptors expressed on myeloid cells (TREMs) encompass a family of cell-surface receptors chiefly expressed by granulocytes, monocytes and tissue macrophages. These receptors have been implicated in inflammation, neurodegenerative diseases, bone remodelling, metabolic syndrome, atherosclerosis and cancer. Here, I review the structure, ligands, signalling modes and functions of TREMs in humans and mice and discuss the challenges that remain in understanding TREM biology.
Collapse
Affiliation(s)
- Marco Colonna
- Department of Pathology and Immunology, Washington University School of Medicine, St Louis, MO, USA.
| |
Collapse
|
5
|
Liu YH, Liu Y, Xin YF, Zhang Q, Ding ML. Identification of key genes involved in calcific aortic valve disease based on integrated bioinformatics analysis. Exp Biol Med (Maywood) 2023; 248:52-60. [PMID: 36151748 PMCID: PMC9989152 DOI: 10.1177/15353702221118088] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
The calcific aortic valve disease (CAVD) develops as an aortic valve sclerosis and progresses to an advanced form of stenosis. In many biological fields, bioinformatics becomes a fundamental component. The key mechanisms involved in CAVD are discovered with the use of bioinformatics to investigate gene function and pathways. We downloaded the original data (GSE51472) from the Gene Expression Omnibus (GEO) database (http://www.ncbi.nlm.nih.gov/geo/). After standardization, 2978 differentially expressed genes (DEGs) were identified from the data sets GSE51472 containing samples from normal, calcified, and sclerotic aortic valves. Analysis of DEGs based on the series test of clusters (STCs) revealed the two most significant patterns. Based on the result of the STC, the functional enrichment analysis of gene ontology (GO) was conducted to investigate the molecular function (MF), biological process (BP), and cell compound (CC) of the DEGs. With a p value of 0.01, DEGs associated with "chronic inflammation," "T-cell receptor complexes," and "antigen binding" had the highest significance within BP, CC, and MF. DEG enrichment in signaling pathways was analyzed using KEGG pathway enrichment. Using a p < 0.05 level of significance, the most enriched biological pathways related to CAVD were "Chemokine signaling pathway," "Cytokine-cytokine receptor interaction," "Tuberculosis," "PI3K-Akt signaling pathway," and "Transcriptional misregulation in cancer." Finally, the construction of gene co-expression networks and pathway networks illustrated the pathogensis of CAVD. TLR2, CD86, and TYROBP were identified as hub genes for the development of CAVD. Moreover, "MAPK signaling pathway," "Apoptosis," and "Pathways in cancer" were regarded as the core pathways among the samples of normal, sclerotic and calcified aortic valve samples.
Collapse
Affiliation(s)
- Ye-Hong Liu
- Department of Cardiology, Shanghai East Hospital, Shanghai Tongji University School of Medicine, Shanghai 200120, China
| | - Yang Liu
- Department of Intensive Care Unit, Shanghai East Hospital, Shanghai Tongji University School of Medicine, Shanghai 200120, China
| | - Yuan-Feng Xin
- Department of Cardiovascular Surgery, Shanghai East Hospital, Shanghai Tongji University School of Medicine, Shanghai 200120, China
| | - Qi Zhang
- Department of Cardiology, Shanghai East Hospital, Shanghai Tongji University School of Medicine, Shanghai 200120, China
| | - Meng-Lei Ding
- Department of Clinical Laboratory, Shanghai East Hospital, Shanghai Tongji University School of Medicine, Shanghai 200120, China
| |
Collapse
|
6
|
Zhang L, Gu J, Wang S, He F, Gong K. Identification of key differential genes in intimal hyperplasia induced by left carotid artery ligation. PeerJ 2022; 10:e13436. [PMID: 35586138 PMCID: PMC9109685 DOI: 10.7717/peerj.13436] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Accepted: 04/22/2022] [Indexed: 01/14/2023] Open
Abstract
Background Intimal hyperplasia is a common pathological process of restenosis following angioplasty, atherosclerosis, pulmonary hypertension, vein graft stenosis, and other proliferative diseases. This study aims to screen for potential novel gene targets and mechanisms related to vascular intimal hyperplasia through an integrated microarray analysis of the Gene Expression Omnibus Database (GEO) database. Material and Methods The gene expression profile of the GSE56143 dataset was downloaded from the Gene Expression Omnibus database. Functional enrichment analysis, protein-protein interaction (PPI) network analysis, and the transcription factor (TF)-target gene regulatory network were used to reveal the biological functions of differential genes (DEGs). Furthermore, the expression levels of the top 10 key DEGs were verified at the mRNA and protein level in the carotid artery 7 days after ligation. Results A total of 373 DEGs (199 upregulated DEGs and 174 downregulated DEGs) were screened. These DEGs were significantly enriched in biological processes, including immune system process, cell adhesion, and several pathways, which were mainly associated with cell adhesion molecules and the regulation of the actin cytoskeleton. The top 10 key DEGs (Ptprc, Fn1, Tyrobp, Emr1, Itgb2, Itgax, CD44, Ctss, Ly86, and Aif1) acted as key genes in the PPI network. The verification of these key DEGs at the mRNA and protein levels was consistent with the results of the above-mentioned bioinformatics analysis. Conclusion The present study identified key genes and pathways involved in intimal hyperplasia induced by carotid artery ligation. These results improved our understanding of the mechanisms underlying the development of intimal hyperplasia and provided candidate targets.
Collapse
Affiliation(s)
- Lina Zhang
- Department of Cardiology, The Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, Jiangsu, China
| | - Jianjun Gu
- Department of Cardiology, Northern Jiangsu People’s Hospital, Yangzhou University, Yangzhou, Jiangsu, China
| | - Sichuan Wang
- Department of Cardiology, The Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, Jiangsu, China
| | - Fuming He
- Department of Cardiology, The Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, Jiangsu, China
| | - Kaizheng Gong
- Department of Cardiology, The Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, Jiangsu, China
| |
Collapse
|
7
|
Luo C, Xia B, Zhong R, Shen D, Li J, Chen L, Zhang H. Early-Life Nutrition Interventions Improved Growth Performance and Intestinal Health via the Gut Microbiota in Piglets. Front Nutr 2022; 8:783688. [PMID: 35047544 PMCID: PMC8762325 DOI: 10.3389/fnut.2021.783688] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2021] [Accepted: 11/29/2021] [Indexed: 12/18/2022] Open
Abstract
Intestinal infections in piglets are the main causes of morbidity before and after weaning. Studies have not explored approaches for combining pre-weaning and post-weaning nutritional strategies to sustain optimal gut health. The current study thus sought to explore the effects of early-life nutrition interventions through administration of synthetic milk on growth performance and gut health in piglets from 3 to 30 days of age. Twelve sows were randomly allocated to control group (CON) and early-life nutrition interventions group (ENI). Piglets were fed with the same creep diet from 7 days of age ad libitum. Piglets in the ENI group were provided with additional synthetic milk from Day 3 to Day 30. The results showed that early-life nutrition interventions improved growth performance, liver weight, spleen weight, and reduced diarrhea rate of piglets after weaning (P < 0.05). Early-life nutrition interventions significantly upregulated expression of ZO-1, Occludin, Claudin4, GALNT1, B3GNT6, and MUC2 in colonic mucosa at mRNA level (P < 0.05). Early-life nutrition interventions reduced activity of alkaline phosphatase (AKP) in serum and the content of lipopolysaccharides (LPS) in plasma (P < 0.05). The number of goblet cells and crypt depth of colon of piglets was significantly higher in piglets in the ENI group relative to that of piglets in the CON group (P < 0.05). The relative mRNA expression levels of MCP-1, TNF-α, IL-1β, and IL-8, and the protein expression levels of TNF-α, IL-6, and IL-8 in colonic mucosa of piglets in the ENI group were lower compared with those of piglets in the CON group (P < 0.05). Relative abundance of Lactobacillus in colonic chyme and mucosa of piglets in the ENI group was significantly higher relative to that of piglets in the CON group (P < 0.05). Correlation analysis indicated that abundance of Lactobacillus was positively correlated with the relative mRNA expression levels of ZO-1, Claudin4, and GALNT1, and it was negatively correlated with the level of MCP-1 in colonic chyme and mucosa. In summary, the findings of this study showed that early-life nutrition interventions improved growth performance, colonic barrier, and reduced inflammation in the colon by modulating composition of gut microbiota in piglets. Early-life nutrition intervention through supplemental synthetic milk is a feasible measure to improve the health and reduce the number of deaths of piglets.
Collapse
Affiliation(s)
- Chengzeng Luo
- State Key Laboratory of Animal Nutrition, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing, China
- College of Animal Science, Xinjiang Agricultural University, Urumqi, China
| | - Bing Xia
- State Key Laboratory of Animal Nutrition, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing, China
- College of Animal Science and Technology, Northwest A&F University, Xianyang, China
| | - Ruqing Zhong
- State Key Laboratory of Animal Nutrition, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Dan Shen
- State Key Laboratory of Animal Nutrition, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Jiaheng Li
- State Key Laboratory of Animal Nutrition, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Liang Chen
- State Key Laboratory of Animal Nutrition, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Hongfu Zhang
- State Key Laboratory of Animal Nutrition, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing, China
| |
Collapse
|
8
|
Lu Z, Tang M, Zhang M, Li Y, Shi F, Zhan F, Zhao L, Li J, Lin L, Qin Z. Hemeprotein amplifies the innate immune receptors of Ctenopharyngodon idellus kidney cells through NF-κB- and MAPK-dependent reactive oxygen species generation. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2022; 126:104207. [PMID: 34273355 DOI: 10.1016/j.dci.2021.104207] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/19/2021] [Revised: 07/13/2021] [Accepted: 07/14/2021] [Indexed: 06/13/2023]
Abstract
Infectious bacterial and viral diseases that cause hemolysis are considered life-threatening to grass carp (Ctenopharyngodon idellus), which is a species used in aquaculture worldwide. After heme and hemeproteins (Hb) are released as a result of hemolysis, the effect of excess Hb and heme on tissues remains to be characterized. To decipher the mechanisms, after incubation with Hb, we showed that lipopolysaccharide (LPS), Hb, and heme increased the cytotoxicity and secretion of inflammatory cytokines such as interleukin (IL)-6, chemokine (C-C motif) ligand 1 (CCL1), tumor necrosis factor (TNF)-α, IL-6, and IL-1β in vitro, which was due to stimulation of the expression of innate immune receptors, such as nucleotide-binding oligomerization domain (NOD2), toll-like receptor 2 (TLR2), TLR 4, and TLR3. The formation of reactive oxygen species (ROS) and the activation of mitogen-activated protein kinases (MAPKs) and nuclear factor (NF)-κB were important for increasing the cytokine production to induce heme, Hb, and LPS. Moreover, we confirmed that after LPS, Hb, and heme challenge, superoxide dismutase (SOD) and glutathione (GSH) synthetase (GSS) also caused remarkable destruction. However, catalase (CAT) and heme oxygenase-1 (HO-1) were strongly activated. In summary, our research findings present a framework through which heme and Hb concentrations amplify the secretions of inflammatory cytokines, which are induced by pattern recognition receptor (PRR) activation and present possible paths for immune intervention during infection with viral diseases and hemolytic bacterial.
Collapse
Affiliation(s)
- Zhijie Lu
- Guangdong Provincial Water Environment and Aquatic Products Security Engineering Technology Research Center, Guangzhou Key Laboratory of Aquatic Animal Diseases and Waterfowl Breeding, College of Animal Sciences and Technology, Zhongkai University of Agriculture and Engineering, Guangzhou, Guangdong Province, 510222, China
| | - Meizhen Tang
- Guangdong Provincial Water Environment and Aquatic Products Security Engineering Technology Research Center, Guangzhou Key Laboratory of Aquatic Animal Diseases and Waterfowl Breeding, College of Animal Sciences and Technology, Zhongkai University of Agriculture and Engineering, Guangzhou, Guangdong Province, 510222, China
| | - Menglan Zhang
- Guangdong Provincial Water Environment and Aquatic Products Security Engineering Technology Research Center, Guangzhou Key Laboratory of Aquatic Animal Diseases and Waterfowl Breeding, College of Animal Sciences and Technology, Zhongkai University of Agriculture and Engineering, Guangzhou, Guangdong Province, 510222, China
| | - Yanan Li
- Guangdong Provincial Water Environment and Aquatic Products Security Engineering Technology Research Center, Guangzhou Key Laboratory of Aquatic Animal Diseases and Waterfowl Breeding, College of Animal Sciences and Technology, Zhongkai University of Agriculture and Engineering, Guangzhou, Guangdong Province, 510222, China
| | - Fei Shi
- Guangdong Provincial Water Environment and Aquatic Products Security Engineering Technology Research Center, Guangzhou Key Laboratory of Aquatic Animal Diseases and Waterfowl Breeding, College of Animal Sciences and Technology, Zhongkai University of Agriculture and Engineering, Guangzhou, Guangdong Province, 510222, China
| | - Fanbin Zhan
- Guangdong Provincial Water Environment and Aquatic Products Security Engineering Technology Research Center, Guangzhou Key Laboratory of Aquatic Animal Diseases and Waterfowl Breeding, College of Animal Sciences and Technology, Zhongkai University of Agriculture and Engineering, Guangzhou, Guangdong Province, 510222, China
| | - Lijuan Zhao
- Guangdong Provincial Water Environment and Aquatic Products Security Engineering Technology Research Center, Guangzhou Key Laboratory of Aquatic Animal Diseases and Waterfowl Breeding, College of Animal Sciences and Technology, Zhongkai University of Agriculture and Engineering, Guangzhou, Guangdong Province, 510222, China
| | - Jun Li
- Guangdong Provincial Water Environment and Aquatic Products Security Engineering Technology Research Center, Guangzhou Key Laboratory of Aquatic Animal Diseases and Waterfowl Breeding, College of Animal Sciences and Technology, Zhongkai University of Agriculture and Engineering, Guangzhou, Guangdong Province, 510222, China; School of Biological Sciences, Lake Superior State University, Sault Ste. Marie, MI, 49783, USA
| | - Li Lin
- Guangdong Provincial Water Environment and Aquatic Products Security Engineering Technology Research Center, Guangzhou Key Laboratory of Aquatic Animal Diseases and Waterfowl Breeding, College of Animal Sciences and Technology, Zhongkai University of Agriculture and Engineering, Guangzhou, Guangdong Province, 510222, China.
| | - Zhendong Qin
- Guangdong Provincial Water Environment and Aquatic Products Security Engineering Technology Research Center, Guangzhou Key Laboratory of Aquatic Animal Diseases and Waterfowl Breeding, College of Animal Sciences and Technology, Zhongkai University of Agriculture and Engineering, Guangzhou, Guangdong Province, 510222, China.
| |
Collapse
|
9
|
Constitutively Activated DAP12 Induces Functional Anti-Tumor Activation and Maturation of Human Monocyte-Derived DC. Int J Mol Sci 2021; 22:ijms22031241. [PMID: 33513928 PMCID: PMC7865632 DOI: 10.3390/ijms22031241] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2021] [Revised: 01/22/2021] [Accepted: 01/22/2021] [Indexed: 01/07/2023] Open
Abstract
Dendritic cells (DCs) are professional antigen presenting cells with a great capacity for cross-presentation of exogenous antigens from which robust anti-tumor immune responses ensue. However, this function is not always available and requires DCs to first be primed to induce their maturation. In particular, in the field of DC vaccine design, currently available methodologies have been limited in eliciting a sustained anti-tumor immune response. Mechanistically, part of the maturation response is influenced by the presence of stimulatory receptors relying on ITAM-containing activating adaptor molecules like DAP12, that modulates their function. We hypothesize that activating DAP12 in DC could force their maturation and enhance their potential anti-tumor activity for therapeutic intervention. For this purpose, we developed constitutively active DAP12 mutants that can promote activation of monocyte-derived DC. Here we demonstrate its ability to induce the maturation and activation of monocyte-derived DCs which enhances migration, and T cell stimulation in vitro using primary human cells. Moreover, constitutively active DAP12 stimulates a strong immune response in a murine melanoma model leading to a reduction of tumor burden. This provides proof-of-concept for investigating the pre-activation of antigen presenting cells to enhance the effectiveness of anti-tumor immunotherapies.
Collapse
|
10
|
Kumar V. Toll-like receptors in sepsis-associated cytokine storm and their endogenous negative regulators as future immunomodulatory targets. Int Immunopharmacol 2020; 89:107087. [PMID: 33075714 PMCID: PMC7550173 DOI: 10.1016/j.intimp.2020.107087] [Citation(s) in RCA: 133] [Impact Index Per Article: 26.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2020] [Revised: 10/04/2020] [Accepted: 10/08/2020] [Indexed: 12/15/2022]
Abstract
Sepsis infects more than 48.9 million people world-wide, with 19.7 million deaths. Cytokine storm plays a significant role in sepsis, along with severe COVID-19. TLR signaling pathways plays a crucial role in generating the cytokine storm. Endogenous negative regulators of TLR signaling are crucial to regulate cytokine storm.
Cytokine storm generates during various systemic acute infections, including sepsis and current pandemic called COVID-19 (severe) causing devastating inflammatory conditions, which include multi-organ failure or multi-organ dysfunction syndrome (MODS) and death of the patient. Toll-like receptors (TLRs) are one of the major pattern recognition receptors (PRRs) expressed by immune cells as well as non-immune cells, including neurons, which play a crucial role in generating cytokine storm. They recognize microbial-associated molecular patterns (MAMPs, expressed by pathogens) and damage or death-associate molecular patterns (DAMPs; released and/expressed by damaged/killed host cells). Upon recognition of MAMPs and DAMPs, TLRs activate downstream signaling pathways releasing several pro-inflammatory mediators [cytokines, chemokines, interferons, and reactive oxygen and nitrogen species (ROS or RNS)], which cause acute inflammation meant to control the pathogen and repair the damage. Induction of an exaggerated response due to genetic makeup of the host and/or persistence of the pathogen due to its evasion mechanisms may lead to severe systemic inflammatory condition called sepsis in response to the generation of cytokine storm and organ dysfunction. The activation of TLR-induced inflammatory response is hardwired to the induction of several negative feedback mechanisms that come into play to conclude the response and maintain immune homeostasis. This state-of-the-art review describes the importance of TLR signaling in the onset of the sepsis-associated cytokine storm and discusses various host-derived endogenous negative regulators of TLR signaling pathways. The subject is very important as there is a vast array of genes and processes implicated in these negative feedback mechanisms. These molecules and mechanisms can be targeted for developing novel therapeutic drugs for cytokine storm-associated diseases, including sepsis, severe COVID-19, and other inflammatory diseases, where TLR-signaling plays a significant role.
Collapse
Affiliation(s)
- V Kumar
- Children Health Clinical Unit, Faculty of Medicine, Mater Research, University of Queensland, ST Lucia, Brisbane, Queensland 4078, Australia; School of Biomedical Sciences, Faculty of Medicine, University of Queensland, ST Lucia, Brisbane, Queensland 4078, Australia.
| |
Collapse
|
11
|
TREML4 receptor regulates inflammation and innate immune cell death during polymicrobial sepsis. Nat Immunol 2020; 21:1585-1596. [DOI: 10.1038/s41590-020-0789-z] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Accepted: 08/21/2020] [Indexed: 12/20/2022]
|
12
|
Dai W, Sun Y, Jiang Z, Du K, Xia N, Zhong G. Key genes associated with non-alcoholic fatty liver disease and acute myocardial infarction. Med Sci Monit 2020; 26:e922492. [PMID: 32594092 PMCID: PMC7341693 DOI: 10.12659/msm.922492] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Background With increasing research on non-alcoholic fatty liver (NAFLD) and acute myocardial infarction (AMI), many studies show a tight correlation between NAFLD and AMI, but the underlying pathophysiology is still not clear. This study was performed to identify the potential hub genes and pathways related to these 2 diseases by using the bioinformatics method. Material/Methods The Gene Expression Omnibus (GEO) dataset GSE63067 of NAFLD patients and normal controls was downloaded from the GEO database. The GSE60993 and GSE66360 datasets for AMI patients and healthy controls were also obtained. Differentially expressed genes (DEGs) of NAFLD and AMI datasets and the common genes between them were obtained. Further GO and KEGG enrichment analyses for common genes were performed. To define the pathogenesis associated with both NAFLD and AMI, a protein–protein interaction (PPI) network was constructed. Finally, SPSS software was utilized to analyze the diagnostic value of hub genes in the NAFLD and AMI datasets, respectively. Results Seventy-eight common genes were obtained in NAFLD and AMI with the threshold of P-value <0.05. Thirty-one GO terms and 10 KEGG pathways were obtained. Also, the top 10 hub genes (TLR2, LILRB2, CXCL1, FPR1, TLR4, TYROBP, MMP9, FCER1G, CLEC4D, and CCR2) were selected with P<0.05. Conclusions The results of this study suggest that some novel genes play an important role in the occurrence and progression NAFLD and AMI. More experimental research and clinical trials are needed to verify our results.
Collapse
Affiliation(s)
- Weiran Dai
- Department of Cardiology Ward 1, The First Affiliated Hospital of Guangxi, Medical University, Guangxi, Nanning, China (mainland)
| | - Yue Sun
- Department of Geriatric Endocrinology, The First Affiliated Hospital of Guangxi, Medical University, Guangxi, Nanning, China (mainland)
| | - Zhiyuan Jiang
- Department of Hypertension, The First Affiliated Hospital of Guangxi, Medical University, Guangxi, Nanning, China (mainland)
| | - Kuan Du
- Department of Geriatric Endocrinology, The First Affiliated Hospital of Guangxi, Medical University, Guangxi, Nanning, China (mainland)
| | - Ning Xia
- Department of Geriatric Endocrinology, The First Affiliated Hospital of Guangxi, Medical University, Guangxi, Nanning, China (mainland)
| | - Guoqiang Zhong
- Department of Cardiology Ward 1, The First Affiliated Hospital of Guangxi, Medical University, Guangxi, Nanning, China (mainland)
| |
Collapse
|
13
|
Chen F, Zhou Y, Wu Z, Li Y, Zhou W, Wang Y. Integrated Analysis of Key Genes and Pathways Involved in Nonalcoholic Steatohepatitis Improvement After Roux-en-Y Gastric Bypass Surgery. Front Endocrinol (Lausanne) 2020; 11:611213. [PMID: 33603714 PMCID: PMC7884850 DOI: 10.3389/fendo.2020.611213] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Accepted: 12/14/2020] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND As the incidence of nonalcoholic fatty liver disease (NAFLD) increases globally, nonalcoholic steatohepatitis (NASH) has become the second common cause of liver transplantation for liver diseases. Recent evidence shows that Roux-en-Y gastric bypass (RYGB) surgery obviously alleviates NASH. However, the mechanism underlying RYGB induced NASH improvement is still elusive. METHODS We obtained datasets, including hepatic gene expression data and histologic NASH status, at baseline and 1 year after RYGB surgery. Differentially expressed genes (DEGs) were identified comparing gene expression before and after RYGB surgery in each dataset. Common DEGs were obtained between both datasets and further subjected to functional and pathway enrichment analysis. Protein-protein interaction (PPI) network was constructed, and key modules and hub genes were also identified. RESULTS In the present study, GSE106737 and GSE83452 datasets were included. One hundred thirty common DEGs (29 up-regulated and 101 down-regulated) were identified between GSE106737 and GSE83452 datasets. KEGG analysis showed that mineral absorption, IL-17 signaling pathway, osteoclast differentiation, and TNF signaling pathway were significantly enriched. Based on the PPI network, IGF1, JUN, FOS, LDLR, TYROBP, DUSP1, CXCR4, ATF3, CXCL2, EGR1, SAA1, CTSS, and PPARA were identified as hub genes, and three functional modules were also extracted. CONCLUSION This study identifies the global gene expression change in the liver of NASH patients before and after RYGB surgery in a bioinformatic method. Our findings will contribute to the understanding of molecular biological changes underlying NASH improvement after RYGB surgery.
Collapse
Affiliation(s)
- Fu Chen
- Department of General Surgery, Fourth Affiliated Hospital of China Medical University, Shenyang, China
| | - Yong Zhou
- Department of General Surgery, Fourth Affiliated Hospital of China Medical University, Shenyang, China
| | - Zhiyuan Wu
- Department of Colorectal and Hernia Minimally Invasive Surgery, Shengjing Hospital of China Medical University, Shenyang, China
| | - Yunze Li
- Department of Colorectal and Hernia Minimally Invasive Surgery, Shengjing Hospital of China Medical University, Shenyang, China
| | - Wenlong Zhou
- Department of General Surgery, The Third Hospital of Shenyang Medical College, Shenyang, China
| | - Yong Wang
- Department of General Surgery, Fourth Affiliated Hospital of China Medical University, Shenyang, China
- *Correspondence: Yong Wang,
| |
Collapse
|
14
|
Nakao T, Ono Y, Dai H, Nakano R, Perez-Gutierrez A, Camirand G, Huang H, Geller DA, Thomson AW. DNAX Activating Protein of 12 kDa/Triggering Receptor Expressed on Myeloid Cells 2 Expression by Mouse and Human Liver Dendritic Cells: Functional Implications and Regulation of Liver Ischemia-Reperfusion Injury. Hepatology 2019; 70:696-710. [PMID: 30372546 PMCID: PMC6488456 DOI: 10.1002/hep.30334] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2018] [Accepted: 10/03/2018] [Indexed: 12/26/2022]
Abstract
Liver interstitial dendritic cells (DCs) have been implicated in the control of ischemia-reperfusion injury (IRI) and host immune responses following liver transplantation. Mechanisms underlying these regulatory functions of hepatic DCs remain unclear. We have shown recently that the transmembrane immunoadaptor DNAX-activating protein of 12 kDa (DAP12) negatively regulates mouse liver DC maturation and proinflammatory and immune stimulatory functions. Here, we used PCR analysis and flow cytometry to characterize expression of DAP12 and its associated triggering receptor, triggering receptor expressed on myeloid cells 2 (TREM2), by mouse and human liver DCs and other immune cells compared with DCs in other tissues. We also examined the roles of DAP12 and TREM2 and their expression by liver DCs in the regulation of liver IRI. Injury was induced in DAP12-/- , TREM2-/- , or wild-type (WT) mice by 1 hour of 70% clamping and quantified following 6 hours of reperfusion. Both DAP12 and TREM2 were coexpressed at comparatively high levels by liver DCs. Mouse liver DCs lacking DAP12 or TREM2 displayed enhanced levels of nuclear factor κB and costimulatory molecule expression. Unlike normal WT liver DCs, DAP12-/- liver DC failed to inhibit proliferative responses of activated T cells. In vivo, DAP12-/- and TREM2-/- mice exhibited enhanced IRI accompanied by augmented liver DC activation. Elevated alanine aminotransferase levels and tissue injury were markedly reduced by infusion of WT but not DAP12-/- DC. Conclusion: Our data reveal a close association between DAP12 and TREM2 expression by liver DC and suggest that, by negatively regulating liver DC stimulatory function, DAP12 promotes their control of hepatic inflammatory responses; the DAP12/TREM2 signaling complex may represent a therapeutic target for control of acute liver injury/liver inflammatory disorders.
Collapse
Affiliation(s)
- Toshimasa Nakao
- Thomas E. Starzl Transplantation Institute, Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Yoshihiro Ono
- Thomas E. Starzl Transplantation Institute, Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Helong Dai
- Thomas E. Starzl Transplantation Institute, Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA,Department of Urological Organ Transplantation, The Second Xiangya Hospital of Central South University, Changsha, P.R. China
| | - Ryosuke Nakano
- Thomas E. Starzl Transplantation Institute, Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Angelica Perez-Gutierrez
- Thomas E. Starzl Transplantation Institute, Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Geoffrey Camirand
- Thomas E. Starzl Transplantation Institute, Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Hai Huang
- Thomas E. Starzl Transplantation Institute, Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - David A. Geller
- Thomas E. Starzl Transplantation Institute, Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA,Liver Cancer Center, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Angus W. Thomson
- Thomas E. Starzl Transplantation Institute, Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA,Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA,Corresponding author: Angus W. Thomson, PhD DSc, Thomas E. Starzl Transplantation Institute, University of Pittsburgh School of Medicine, 200 Lothrop Street, W1540 BST, Pittsburgh, PA 15261, Phone: (412) 624-6392, Fax: (412)-624-1172,
| |
Collapse
|
15
|
Rojas MA, Shen ZT, Caldwell RB, Sigalov AB. Blockade of TREM-1 prevents vitreoretinal neovascularization in mice with oxygen-induced retinopathy. Biochim Biophys Acta Mol Basis Dis 2018; 1864:2761-2768. [PMID: 29730341 PMCID: PMC6488934 DOI: 10.1016/j.bbadis.2018.05.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2018] [Revised: 04/25/2018] [Accepted: 05/01/2018] [Indexed: 12/20/2022]
Abstract
In pathological retinal neovascularization (RNV) disorders, the retina is infiltrated by activated leukocytes and macrophages. Triggering receptor expressed on myeloid cells 1 (TREM-1), an inflammation amplifier, activates monocytes and macrophages and plays an important role in cancer, autoimmune and other inflammation-associated disorders. Hypoxia-inducible TREM-1 is involved in cancer angiogenesis but its role in RNV remains unclear. Here, to close this gap, we evaluated the role of TREM-1 in RNV using a mouse model of oxygen-induced retinopathy (OIR). We found that hypoxia induced overexpression of TREM-1 in the OIR retinas compared to that of the room air group. TREM-1 was observed specifically in areas of pathological RNV, largely colocalizing with macrophage colony-stimulating factor (M-CSF) and CD45- and Iba-1-positive cells. TREM-1 blockade using systemically administered first-in-class ligand-independent TREM-1 inhibitory peptides rationally designed using the signaling chain homooligomerization (SCHOOL) strategy significantly (up to 95%) reduced vitreoretinal neovascularization. The peptides were well-tolerated when formulated into lipopeptide complexes for peptide half-life extension and targeted delivery. TREM-1 inhibition substantially downregulated retinal protein levels of TREM-1 and M-CSF suggesting that TREM-1-dependent suppression of pathological angiogenesis involves M-CSF. Targeting TREM-1 using TREM-1-specific SCHOOL peptide inhibitors represents a novel strategy to treat retinal diseases that are accompanied by neovascularization including retinopathy of prematurity.
Collapse
Affiliation(s)
- Modesto A Rojas
- Vascular Biology Center, Augusta University, Augusta, GA 30912, United States.
| | - Zu T Shen
- SignaBlok, Inc, P.O. Box 4064, Shrewsbury, MA 01545, United States
| | - Ruth B Caldwell
- Vascular Biology Center, Augusta University, Augusta, GA 30912, United States; Charlie Norwood VA Medical Center, Augusta, GA 30904, United States
| | | |
Collapse
|
16
|
TREM2/DAP12 Signal Elicits Proinflammatory Response in Microglia and Exacerbates Neuropathic Pain. J Neurosci 2017; 36:11138-11150. [PMID: 27798193 DOI: 10.1523/jneurosci.1238-16.2016] [Citation(s) in RCA: 106] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2016] [Accepted: 09/12/2016] [Indexed: 11/21/2022] Open
Abstract
Neuropathic pain afflicts millions of people, and the development of an effective treatment for this intractable pain is an urgent issue. Recent evidence has implicated microglia in neuropathic pain. The present study showed that the DNAX-activating protein of 12 kDa (DAP12) and its associated "triggering receptor expressed on myeloid cells 2" (TREM2) were predominantly expressed by microglia in the dorsal horn after spinal nerve injury, revealing a role for TREM2/DAP12 signaling in neuropathic pain. Nerve injury-induced proinflammatory cytokine expression in microglia and pain behaviors were significantly suppressed in Dap12-deficient mice. Furthermore, intrathecal administration of TREM2 agonistic antibody induced proinflammatory cytokine expression, as well as neuropathic pain, in mice without nerve injury. The agonistic antibody induced proinflammatory responses and neuropathic pain was not observed in Dap12-deficient mice. Together, these results suggest that TREM2/DAP12-mediated signals in microglia exacerbate nerve injury-induced neuropathic pain by inducing proinflammatory cytokine secretion from microglia. Suppression of DAP12-mediated signals could be a therapeutic target for neuropathic pain. SIGNIFICANCE STATEMENT Recent studies have revealed that activated microglia in the spinal dorsal horn exacerbate neuropathic pain, which has suggested that suppression of microglial activity should be considered as a therapeutic target. However, only a few molecules have been identified as regulators of microglial activity. In this study, we focused on a receptor complex of TREM2 and DAP12, both of which are expressed by microglia and have been implicated in the pathogenesis of Alzheimer's disease, and demonstrated that TREM2/DAP12 signaling promoted proinflammatory responses in microglia and exacerbates neuropathic pain. The present results revealed the functional significance of TREM2/DAP12 signaling in microglial activation after neuronal injury, and could help in the development of treatments for neuropathic pain and neurodegenerative diseases.
Collapse
|
17
|
Zhong L, Zhang ZL, Li X, Liao C, Mou P, Wang T, Wang Z, Wang Z, Wei M, Xu H, Bu G, Chen XF. TREM2/DAP12 Complex Regulates Inflammatory Responses in Microglia via the JNK Signaling Pathway. Front Aging Neurosci 2017; 9:204. [PMID: 28680398 PMCID: PMC5478682 DOI: 10.3389/fnagi.2017.00204] [Citation(s) in RCA: 59] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2017] [Accepted: 06/06/2017] [Indexed: 12/26/2022] Open
Abstract
DNAX-activating protein of 12 kDa (DAP12) is a signaling adapter protein expressed in cells that participate in innate immune responses. By pairing with different triggering receptors expressed on myeloid cell (TREM) proteins, DAP12 can mediate both positive and negative cellular responses. In particular, TREM1 acts as an amplifier of the immune response, while TREM2 functions as a negative regulator. TREM2 has also been shown to stimulate the phagocytosis of apoptotic neurons and define the barrier function in microglia. Notably, loss-of-function mutations of either DAP12 or TREM2 result in a disorder known as Nasu-Hakola disease (NHD); and mutations of these genes have been associated with the risk for Alzheimer's disease (AD), suggesting that TREM2 and DAP12 may regulate common signaling pathways in the disease pathogenesis. In this study, we demonstrated an anti-inflammatory role of DAP12 in murine microglia that depends on the presence of TREM2. We also uncovered the JNK signaling pathway as the underlying molecular mechanism by which the TREM2/DAP12 complex suppresses the hyperactivation of microglia upon LPS stimulation. Interestingly, LPS down-regulates the expression of Trem2 via the activation of JNK and NF-κB signaling pathways, resulting in a vicious cycle that synergistically promotes the inflammatory responses. Our study provides insights into mechanism-based therapy for neuroinflammatory disorders.
Collapse
Affiliation(s)
- Li Zhong
- Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, Medical College, Xiamen UniversityXiamen, China
| | - Zhen-Lian Zhang
- Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, Medical College, Xiamen UniversityXiamen, China
| | - Xinxiu Li
- Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, Medical College, Xiamen UniversityXiamen, China
| | - Chunyan Liao
- Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, Medical College, Xiamen UniversityXiamen, China
| | - Pengfei Mou
- Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, Medical College, Xiamen UniversityXiamen, China
| | - Tingting Wang
- Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, Medical College, Xiamen UniversityXiamen, China
| | - Zongqi Wang
- Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, Medical College, Xiamen UniversityXiamen, China
| | - Zhe Wang
- Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, Medical College, Xiamen UniversityXiamen, China
| | - Min Wei
- Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, Medical College, Xiamen UniversityXiamen, China
| | - Huaxi Xu
- Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, Medical College, Xiamen UniversityXiamen, China.,Neuroscience and Aging Research Center, Sanford-Burnham-Prebys Medical Discovery InstituteLa Jolla, CA, United States
| | - Guojun Bu
- Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, Medical College, Xiamen UniversityXiamen, China.,Department of Neuroscience, Mayo ClinicJacksonville, FL, United States
| | - Xiao-Fen Chen
- Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, Medical College, Xiamen UniversityXiamen, China.,Shenzhen Research Institute of Xiamen UniversityShenzhen, China
| |
Collapse
|
18
|
Voss OH, Murakami Y, Pena MY, Lee HN, Tian L, Margulies DH, Street JM, Yuen PST, Qi CF, Krzewski K, Coligan JE. Lipopolysaccharide-Induced CD300b Receptor Binding to Toll-like Receptor 4 Alters Signaling to Drive Cytokine Responses that Enhance Septic Shock. Immunity 2016; 44:1365-78. [PMID: 27261276 DOI: 10.1016/j.immuni.2016.05.005] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2015] [Revised: 02/03/2016] [Accepted: 02/20/2016] [Indexed: 12/12/2022]
Abstract
Receptor CD300b is implicated in regulating the immune response to bacterial infection by an unknown mechanism. Here, we identified CD300b as a lipopolysaccharide (LPS)-binding receptor and determined the mechanism underlying CD300b augmentation of septic shock. In vivo depletion and adoptive transfer studies identified CD300b-expressing macrophages as the key cell type augmenting sepsis. We showed that CD300b, and its adaptor DAP12, associated with Toll-like receptor 4 (TLR4) upon LPS binding, thereby enhancing TLR4-adaptor MyD88- and TRIF-dependent signaling that resulted in an elevated pro-inflammatory cytokine storm. LPS engagement of the CD300b-TLR4 complex led to the recruitment and activation of spleen tyrosine kinase (Syk) and phosphatidylinositol-4,5-bisphosphate 3-kinase (PI3K). This resulted in an inhibition of the ERK1/2 protein kinase- and NF-κB transcription factor-mediated signaling pathways, which subsequently led to a reduced interleukin-10 (IL-10) production. Collectively, our data describe a mechanism of TLR4 signaling regulated by CD300b in myeloid cells in response to LPS.
Collapse
Affiliation(s)
- Oliver H Voss
- Receptor Cell Biology Section, Laboratory of Immunogenetics, National Institute of Allergy and Infectious Diseases, NIH, Rockville, MD 20852, USA
| | - Yousuke Murakami
- Receptor Cell Biology Section, Laboratory of Immunogenetics, National Institute of Allergy and Infectious Diseases, NIH, Rockville, MD 20852, USA
| | - Mirna Y Pena
- Receptor Cell Biology Section, Laboratory of Immunogenetics, National Institute of Allergy and Infectious Diseases, NIH, Rockville, MD 20852, USA
| | - Ha-Na Lee
- Receptor Cell Biology Section, Laboratory of Immunogenetics, National Institute of Allergy and Infectious Diseases, NIH, Rockville, MD 20852, USA
| | - Linjie Tian
- Receptor Cell Biology Section, Laboratory of Immunogenetics, National Institute of Allergy and Infectious Diseases, NIH, Rockville, MD 20852, USA
| | - David H Margulies
- Molecular Biology Section, Laboratory of Immunology, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, MD 20814, USA
| | - Jonathan M Street
- Renal Diagnostics and Therapeutics Unit, National Institute of Diabetes and Digestive and Kidney Diseases, NIH, Bethesda, MD 20814, USA
| | - Peter S T Yuen
- Renal Diagnostics and Therapeutics Unit, National Institute of Diabetes and Digestive and Kidney Diseases, NIH, Bethesda, MD 20814, USA
| | - Chen-Feng Qi
- Pathology Core, Laboratory of Immunogenetics, National Institute of Allergy and Infectious Diseases, NIH, Rockville, MD 20852, USA
| | - Konrad Krzewski
- Receptor Cell Biology Section, Laboratory of Immunogenetics, National Institute of Allergy and Infectious Diseases, NIH, Rockville, MD 20852, USA
| | - John E Coligan
- Receptor Cell Biology Section, Laboratory of Immunogenetics, National Institute of Allergy and Infectious Diseases, NIH, Rockville, MD 20852, USA.
| |
Collapse
|
19
|
Wang J, Wei B, Cao S, Xu F, Chen W, Lin H, Du C, Sun Z. Identification by microarray technology of key genes involved in the progression of carotid atherosclerotic plaque. Genes Genet Syst 2015; 89:253-8. [PMID: 25948119 DOI: 10.1266/ggs.89.253] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
A comparative analysis of gene expression profiles between early and advanced carotid atherosclerotic plaque was performed to identify key genes and pathways involved in the progression of carotid atherosclerotic plaque. Gene expression data set GSE28829 was downloaded from Gene Expression Omnibus, including 13 early and 16 advanced atherosclerotic plaque samples from human carotid. Differentially expressed genes (DEGs) were identified using the package limma of R. Principal component analysis was carried out for the DEGs with package rgl of R. A gene coexpression network was constructed with information from COXPRESdb and then visualized with Cytoscape. Functional enrichment analysis was performed with DAVID and pathway enrichment analysis was done with KEGG. A total of 319 DEGs were identified in the advanced atherosclerotic plaque samples compared with early atherosclerotic plaque samples, including 267 up-regulated genes and 52 down-regulated genes. In the gene coexpression network, TYRO protein tyrosine kinase binding protein was the hub gene with a degree of 23. Functional enrichment analysis and pathway enrichment analysis suggested that the immune response played a critical role in the progression of carotid atherosclerotic plaque. A number of key genes were revealed in carotid atherosclerotic plaque, and are potential biomarkers for diagnosis or treatment. These findings may also guide future research to better decipher the progression of atherosclerosis.
Collapse
Affiliation(s)
- Jian Wang
- 1Department of Neurosurgery, China-Japan Union Hospital of Jilin University, Changchun, Jilin, 130033; 2. Department of Neurosurgery, The Affiliated Hospital of Inner Mongolia National University, Tongliao, Inner Mongolia, 028000, China
| | | | | | | | | | | | | | | |
Collapse
|
20
|
Microglia Function in Central Nervous System Development and Plasticity. Cold Spring Harb Perspect Biol 2015; 7:a020545. [PMID: 26187728 DOI: 10.1101/cshperspect.a020545] [Citation(s) in RCA: 252] [Impact Index Per Article: 25.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
The nervous system comprises a remarkably diverse and complex network of different cell types, which must communicate with one another with speed, reliability, and precision. Thus, the developmental patterning and maintenance of these cell populations and their connections with one another pose a rather formidable task. Emerging data implicate microglia, the resident myeloid-derived cells of the central nervous system (CNS), in the spatial patterning and synaptic wiring throughout the healthy, developing, and adult CNS. Importantly, new tools to specifically manipulate microglia function have revealed that these cellular functions translate, on a systems level, to effects on overall behavior. In this review, we give a historical perspective of work to identify microglia function in the healthy CNS and highlight exciting new work in the field that has identified roles for these cells in CNS development, maintenance, and plasticity.
Collapse
|
21
|
Zhong L, Chen XF, Zhang ZL, Wang Z, Shi XZ, Xu K, Zhang YW, Xu H, Bu G. DAP12 Stabilizes the C-terminal Fragment of the Triggering Receptor Expressed on Myeloid Cells-2 (TREM2) and Protects against LPS-induced Pro-inflammatory Response. J Biol Chem 2015; 290:15866-15877. [PMID: 25957402 DOI: 10.1074/jbc.m115.645986] [Citation(s) in RCA: 112] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2015] [Indexed: 11/06/2022] Open
Abstract
Triggering receptor expressed on myeloid cells 2 (TREM2) is a DAP12-associated receptor expressed in microglia, macrophages, and other myeloid-derived cells. Previous studies have suggested that TREM2/DAP12 signaling pathway reduces inflammatory responses and promotes phagocytosis of apoptotic neurons. Recently, TREM2 has been identified as a risk gene for Alzheimer disease (AD). Here, we show that DAP12 stabilizes the C-terminal fragment of TREM2 (TREM2-CTF), a substrate for γ-secretase. Co-expression of DAP12 with TREM2 selectively increased the level of TREM2-CTF with little effects on that of full-length TREM2. The interaction between DAP12 and TREM2 is essential for TREM2-CTF stabilization as a mutant form of DAP12 with disrupted interaction with TREM2 failed to exhibit such an effect. Silencing of either Trem2 or Dap12 gene significantly exacerbated pro-inflammatory responses induced by lipopolysaccharides (LPS). Importantly, overexpression of either full-length TREM2 or TREM2-CTF reduced LPS-induced inflammatory responses. Taken together, our results support a role of DAP12 in stabilizing TREM2-CTF, thereby protecting against excessive pro-inflammatory responses.
Collapse
Affiliation(s)
- Li Zhong
- Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, Medical College, Xiamen University, Xiamen 361102, PR China
| | - Xiao-Fen Chen
- Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, Medical College, Xiamen University, Xiamen 361102, PR China.
| | - Zhen-Lian Zhang
- Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, Medical College, Xiamen University, Xiamen 361102, PR China
| | - Zhe Wang
- Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, Medical College, Xiamen University, Xiamen 361102, PR China
| | - Xin-Zhen Shi
- Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, Medical College, Xiamen University, Xiamen 361102, PR China
| | - Kai Xu
- Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, Medical College, Xiamen University, Xiamen 361102, PR China
| | - Yun-Wu Zhang
- Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, Medical College, Xiamen University, Xiamen 361102, PR China
| | - Huaxi Xu
- Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, Medical College, Xiamen University, Xiamen 361102, PR China; Degenerative Disease Research Program, Center for Neuroscience, Aging, and Stem Cell Research, Sanford-Burnham Medical Research Institute, La Jolla, California 92037
| | - Guojun Bu
- Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, Medical College, Xiamen University, Xiamen 361102, PR China; Department of Neuroscience, Mayo Clinic, Jacksonville, Florida 32224.
| |
Collapse
|
22
|
Spahn JH, Li W, Bribriesco AC, Liu J, Shen H, Ibricevic A, Pan JH, Zinselmeyer BH, Brody SL, Goldstein DR, Krupnick AS, Gelman AE, Miller MJ, Kreisel D. DAP12 expression in lung macrophages mediates ischemia/reperfusion injury by promoting neutrophil extravasation. THE JOURNAL OF IMMUNOLOGY 2015; 194:4039-48. [PMID: 25762783 DOI: 10.4049/jimmunol.1401415] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/02/2014] [Accepted: 02/04/2015] [Indexed: 12/25/2022]
Abstract
Neutrophils are critical mediators of innate immune responses and contribute to tissue injury. However, immune pathways that regulate neutrophil recruitment to injured tissues during noninfectious inflammation remain poorly understood. DAP12 is a cell membrane-associated protein that is expressed in myeloid cells and can either augment or dampen innate inflammatory responses during infections. To elucidate the role of DAP12 in pulmonary ischemia/reperfusion injury (IRI), we took advantage of a clinically relevant mouse model of transplant-mediated lung IRI. This technique allowed us to dissect the importance of DAP12 in tissue-resident cells and those that infiltrate injured tissue from the periphery during noninfectious inflammation. Macrophages in both mouse and human lungs that have been subjected to cold ischemic storage express DAP12. We found that donor, but not recipient, deficiency in DAP12 protected against pulmonary IRI. Analysis of the immune response showed that DAP12 promotes the survival of tissue-resident alveolar macrophages and contributes to local production of neutrophil chemoattractants. Intravital imaging demonstrated a transendothelial migration defect into DAP12-deficient lungs, which can be rescued by local administration of the neutrophil chemokine CXCL2. We have uncovered a previously unrecognized role for DAP12 expression in tissue-resident alveolar macrophages in mediating acute noninfectious tissue injury through regulation of neutrophil trafficking.
Collapse
Affiliation(s)
- Jessica H Spahn
- Department of Surgery, Washington University, St. Louis, MO 63110
| | - Wenjun Li
- Department of Surgery, Washington University, St. Louis, MO 63110
| | | | - Jie Liu
- Department of Surgery, Washington University, St. Louis, MO 63110
| | - Hua Shen
- Department of Internal Medicine, Yale School of Medicine, New Haven, CT 06510; Department of Immunobiology, Yale School of Medicine, New Haven, CT 06510
| | - Aida Ibricevic
- Department of Medicine, Washington University, St. Louis, MO 63110; and
| | - Jie-Hong Pan
- Department of Medicine, Washington University, St. Louis, MO 63110; and
| | - Bernd H Zinselmeyer
- Department of Pathology & Immunology, Washington University, St. Louis, MO 63110
| | - Steven L Brody
- Department of Medicine, Washington University, St. Louis, MO 63110; and
| | - Daniel R Goldstein
- Department of Internal Medicine, Yale School of Medicine, New Haven, CT 06510; Department of Immunobiology, Yale School of Medicine, New Haven, CT 06510
| | | | - Andrew E Gelman
- Department of Surgery, Washington University, St. Louis, MO 63110; Department of Pathology & Immunology, Washington University, St. Louis, MO 63110
| | - Mark J Miller
- Department of Medicine, Washington University, St. Louis, MO 63110; and
| | - Daniel Kreisel
- Department of Surgery, Washington University, St. Louis, MO 63110; Department of Pathology & Immunology, Washington University, St. Louis, MO 63110
| |
Collapse
|
23
|
Kobayashi M, Konishi H, Takai T, Kiyama H. A DAP12-dependent signal promotes pro-inflammatory polarization in microglia following nerve injury and exacerbates degeneration of injured neurons. Glia 2015; 63:1073-82. [PMID: 25690660 DOI: 10.1002/glia.22802] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2014] [Accepted: 01/16/2015] [Indexed: 01/17/2023]
Abstract
Under pathological conditions, activated microglia play paradoxical roles and could have neurotoxic or neuroprotective effects. However, the signal determining how activated microglia affects the fate of neuronal cells remains largely unknown. Here we demonstrate that DNAX-activating protein of 12 kDa (DAP12), a transmembrane adaptor protein that contains an immunoreceptor tyrosine-based activation motif, is a critical regulator of microglial function after nerve injury. In a model of mouse hypoglossal nerve injury, the duration of microglial increase after nerve injury became shorter in mice lacking DAP12, although microglial morphology and total cell numbers were not significantly affected during early phase after nerve injury. Intriguingly, expressions of M1-phenotype markers including pro-inflammatory cytokines were suppressed in DAP12-deficient microglia. Furthermore, axotomy-induced motor neuron death was markedly prevented in DAP12-deficient mice. Collectively, DAP12-mediated microglial activation following axotomy promotes pro-inflammatory responses, and thereby accelerates nerve injury-induced neuron death, suggesting that DAP12 is a potential therapeutic target for the protection of neuronal degeneration caused by microglial activation.
Collapse
Affiliation(s)
- Masaaki Kobayashi
- Department of Functional Anatomy and Neuroscience, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | | | | | | |
Collapse
|
24
|
Abstract
OBJECTIVES Streptococcus pneumoniae is the most common causative organism in community-acquired pneumonia responsible for millions of deaths every year. DNAX-activating protein of 12 kDa is an adaptor molecule for different myeloid expressed receptors involved in innate immunity. DESIGN Animal study. SETTING University research laboratory. SUBJECTS DNAX-activating protein of 12 kDa-deficient (dap12) and wild-type mice. INTERVENTIONS Mice were intranasally infected with S. pneumoniae. In addition, ex vivo responsiveness of alveolar macrophages was examined. MEASUREMENTS AND MAIN RESULTS dap12 alveolar macrophages released more tumor necrosis factor-α upon stimulation with S. pneumoniae and displayed increased phagocytosis of this pathogen compared with wild-type cells. After infection with S. pneumoniae via the airways, dap12 mice demonstrated reduced bacterial outgrowth in the lungs together with delayed dissemination to distant body sites relative to wild-type mice. This favorable response in dap12 mice was accompanied by reduced lung inflammation and an improved survival. CONCLUSIONS These data suggest that DNAX-activating protein of 12 kDa impairs host defense during pneumococcal pneumonia at the primary site of infection at least in part by inhibiting phagocytosis by alveolar macrophages.
Collapse
|
25
|
Dutra FF, Bozza MT. Heme on innate immunity and inflammation. Front Pharmacol 2014; 5:115. [PMID: 24904418 PMCID: PMC4035012 DOI: 10.3389/fphar.2014.00115] [Citation(s) in RCA: 235] [Impact Index Per Article: 21.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2014] [Accepted: 04/29/2014] [Indexed: 12/30/2022] Open
Abstract
Heme is an essential molecule expressed ubiquitously all through our tissues. Heme plays major functions in cellular physiology and metabolism as the prosthetic group of diverse proteins. Once released from cells and from hemeproteins free heme causes oxidative damage and inflammation, thus acting as a prototypic damage-associated molecular pattern. In this context, free heme is a critical component of the pathological process of sterile and infectious hemolytic conditions including malaria, hemolytic anemias, ischemia-reperfusion, and hemorrhage. The plasma scavenger proteins hemopexin and albumin reduce heme toxicity and are responsible for transporting free heme to intracellular compartments where it is catabolized by heme-oxygenase enzymes. Upon hemolysis or severe cellular damage the serum capacity to scavenge heme may saturate and increase free heme to sufficient amounts to cause tissue damage in various organs. The mechanism by which heme causes reactive oxygen generation, activation of cells of the innate immune system and cell death are not fully understood. Although heme can directly promote lipid peroxidation by its iron atom, heme can also induce reactive oxygen species generation and production of inflammatory mediators through the activation of selective signaling pathways. Heme activates innate immune cells such as macrophages and neutrophils through activation of innate immune receptors. The importance of these events has been demonstrated in infectious and non-infectious diseases models. In this review, we will discuss the mechanisms behind heme-induced cytotoxicity and inflammation and the consequences of these events on different tissues and diseases.
Collapse
Affiliation(s)
- Fabianno F. Dutra
- Laboratório de Inflamação e Imunidade, Departamento de Imunologia, Instituto de Microbiologia, Universidade Federal do Rio de JaneiroRio de Janeiro, Brazil
| | - Marcelo T. Bozza
- Laboratório de Inflamação e Imunidade, Departamento de Imunologia, Instituto de Microbiologia, Universidade Federal do Rio de JaneiroRio de Janeiro, Brazil
| |
Collapse
|
26
|
Montalvo V, Quigley L, Vistica BP, Boelte KC, Nugent LF, Takai T, McVicar DW, Gery I. Environmental factors determine DAP12 deficiency to either enhance or suppress immunopathogenic processes. Immunology 2014; 140:475-82. [PMID: 23906311 DOI: 10.1111/imm.12158] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2013] [Revised: 06/28/2013] [Accepted: 07/29/2013] [Indexed: 12/30/2022] Open
Abstract
DNAX-activation protein 12 (DAP12), a transmembrane adapter, plays a major role in transducing activation signals in natural killer cells and various myeloid cells. Quantitative RT-PCR detected in normal mouse eyes considerable levels of DAP12 and multiple DAP12-coupled receptors, in particular TREM-1, Clec5a and SIRPb1. The role of DAP12 and its receptors in experimental autoimmune diseases has been controversial. Here, we analysed the effect of DAP12 deficiency on the capacity of mice to mount immunopathogenic cellular responses to the uveitogenic ocular antigen and interphotoreceptor retinoid-binding protein (IRBP), and to develop experimental autoimmune uveitis (EAU). Surprisingly, sequential analysis of EAU in mice deficient in DAP12 in two different animal facilities at first revealed enhanced disease as compared with wild-type mice, but when these mice were re-derived into a second, cleaner, animal facility, the response of control mice was essentially unchanged, whereas the DAP12 null mice were markedly hyporesponsive relative to controls in the new facility. Accordingly, when stimulated in vitro with IRBP, lymphocytes from the DAP12-deficient mice housed in the two facilities proliferated and produced opposite profiles of pro-inflammatory and anti-inflammatory cytokines, compared with their controls. These findings therefore demonstrate that the effects of DAP12 deficiency on development of autoimmune disease are dramatically affected by environmental factors.
Collapse
Affiliation(s)
- Vanessa Montalvo
- Laboratory of Immunology, National Eye Institute, NIH, Bethesda, MD, USA
| | | | | | | | | | | | | | | |
Collapse
|
27
|
Weber B, Schuster S, Zysset D, Rihs S, Dickgreber N, Schürch C, Riether C, Siegrist M, Schneider C, Pawelski H, Gurzeler U, Ziltener P, Genitsch V, Tacchini-Cottier F, Ochsenbein A, Hofstetter W, Kopf M, Kaufmann T, Oxenius A, Reith W, Saurer L, Mueller C. TREM-1 deficiency can attenuate disease severity without affecting pathogen clearance. PLoS Pathog 2014; 10:e1003900. [PMID: 24453980 PMCID: PMC3894224 DOI: 10.1371/journal.ppat.1003900] [Citation(s) in RCA: 112] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2013] [Accepted: 12/10/2013] [Indexed: 12/02/2022] Open
Abstract
Triggering receptor expressed on myeloid cells-1 (TREM-1) is a potent amplifier of pro-inflammatory innate immune reactions. While TREM-1-amplified responses likely aid an improved detection and elimination of pathogens, excessive production of cytokines and oxygen radicals can also severely harm the host. Studies addressing the pathogenic role of TREM-1 during endotoxin-induced shock or microbial sepsis have so far mostly relied on the administration of TREM-1 fusion proteins or peptides representing part of the extracellular domain of TREM-1. However, binding of these agents to the yet unidentified TREM-1 ligand could also impact signaling through alternative receptors. More importantly, controversial results have been obtained regarding the requirement of TREM-1 for microbial control. To unambiguously investigate the role of TREM-1 in homeostasis and disease, we have generated mice deficient in Trem1. Trem1−/− mice are viable, fertile and show no altered hematopoietic compartment. In CD4+ T cell- and dextran sodium sulfate-induced models of colitis, Trem1−/− mice displayed significantly attenuated disease that was associated with reduced inflammatory infiltrates and diminished expression of pro-inflammatory cytokines. Trem1−/− mice also exhibited reduced neutrophilic infiltration and decreased lesion size upon infection with Leishmania major. Furthermore, reduced morbidity was observed for influenza virus-infected Trem1−/− mice. Importantly, while immune-associated pathologies were significantly reduced, Trem1−/− mice were equally capable of controlling infections with L. major, influenza virus, but also Legionella pneumophila as Trem1+/+ controls. Our results not only demonstrate an unanticipated pathogenic impact of TREM-1 during a viral and parasitic infection, but also indicate that therapeutic blocking of TREM-1 in distinct inflammatory disorders holds considerable promise by blunting excessive inflammation while preserving the capacity for microbial control. Triggering receptor expressed on myeloid cells-1 (TREM-1) is an immune receptor expressed by myeloid cells that has the capacity to augment pro-inflammatory responses in the context of a microbial infection. While a TREM-1-amplified response likely serves the efficient clearance of pathogens, it also bears the potential to cause substantial tissue damage or even death. Hence, TREM-1 appears a possible therapeutic target for tempering deleterious host-pathogen interactions. However, in models of bacterial sepsis controversial findings have been obtained regarding the requirement of TREM-1 for bacterial control - depending on the overall degree of the TREM-1 blockade that was achieved. In order to conclusively investigate harmful versus essential functions of TREM-1 in vivo, we have generated mice deficient in Trem1. Trem1−/− mice were subjected to experimentally-induced intestinal inflammation (as a model of a non-infectious, yet microbial-driven disease) and also analysed following infections with Leishmania major, influenza virus and Legionella pneumophila. Across all models analysed, Trem1−/− mice showed substantially reduced immune-associated disease. We thus describe a previously unanticipated pathogenic role for TREM-1 also during a parasitic and viral infection. Importantly, our data suggest that in certain diseases microbial control can be achieved in the context of blunted inflammation in the absence of TREM-1.
Collapse
Affiliation(s)
- Benjamin Weber
- Division of Experimental Pathology, Institute of Pathology, University of Bern, Bern, Switzerland
| | - Steffen Schuster
- Department of Biochemistry, University of Lausanne, Epalinges, Switzerland
| | - Daniel Zysset
- Division of Experimental Pathology, Institute of Pathology, University of Bern, Bern, Switzerland
| | - Silvia Rihs
- Division of Experimental Pathology, Institute of Pathology, University of Bern, Bern, Switzerland
| | - Nina Dickgreber
- Division of Experimental Pathology, Institute of Pathology, University of Bern, Bern, Switzerland
| | - Christian Schürch
- Division of Experimental Pathology, Institute of Pathology, University of Bern, Bern, Switzerland
- Department of Clinical Research, University of Bern, Bern, Switzerland
| | - Carsten Riether
- Department of Clinical Research, University of Bern, Bern, Switzerland
| | - Mark Siegrist
- Department of Clinical Research, University of Bern, Bern, Switzerland
| | | | - Helga Pawelski
- Institute of Molecular Health Sciences, ETH Zurich, Zurich, Switzerland
| | - Ursina Gurzeler
- Institute of Pharmacology, University of Bern, Bern, Switzerland
| | | | - Vera Genitsch
- Division of Experimental Pathology, Institute of Pathology, University of Bern, Bern, Switzerland
| | | | - Adrian Ochsenbein
- Department of Clinical Research, University of Bern, Bern, Switzerland
- Department of Medical Oncology, University of Bern, Bern, Switzerland
| | - Willy Hofstetter
- Department of Clinical Research, University of Bern, Bern, Switzerland
| | - Manfred Kopf
- Institute of Molecular Health Sciences, ETH Zurich, Zurich, Switzerland
| | - Thomas Kaufmann
- Institute of Pharmacology, University of Bern, Bern, Switzerland
| | | | - Walter Reith
- Department of Pathology and Immunology, Centre Medical Universitaire, Geneva, Switzerland
| | - Leslie Saurer
- Division of Experimental Pathology, Institute of Pathology, University of Bern, Bern, Switzerland
- * E-mail: (LS); (CM)
| | - Christoph Mueller
- Division of Experimental Pathology, Institute of Pathology, University of Bern, Bern, Switzerland
- * E-mail: (LS); (CM)
| |
Collapse
|
28
|
Pelham CJ, Agrawal DK. Emerging roles for triggering receptor expressed on myeloid cells receptor family signaling in inflammatory diseases. Expert Rev Clin Immunol 2013; 10:243-56. [PMID: 24325404 DOI: 10.1586/1744666x.2014.866519] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Innate immune receptors represent important therapeutic targets for inflammatory disorders. In particular, the Toll-like receptor (TLR) family has emerged as a promoter of chronic inflammation that contributes to obesity, insulin resistance and atherosclerosis. Importantly, triggering receptor expressed on myeloid cells-1 (TREM-1) has been characterized as an 'amplifier' of TLR2 and TLR4 signaling. TREM-1- and TREM-2-dependent signaling, as opposed to TREM-like transcript-1 (TLT-1 or TREML1), are mediated through association with the transmembrane adaptor DNAX activation protein of 12 kDa (DAP12). Recessive inheritance of rare mutations in DAP12 or TREM-2 results in a disorder called polycystic lipomembranous osteodysplasia with sclerosing leukoencephalopathy, and surprisingly these subjects are not immunocompromised. Recent progress into the roles of TREM/DAP12 signaling is critically reviewed here with a focus on metabolic, cardiovascular and inflammatory diseases. The expanding repertoire of putative ligands for TREM receptors is also discussed.
Collapse
Affiliation(s)
- Christopher J Pelham
- Department of Biomedical Sciences and Center for Clinical & Translational Science, Creighton University School of Medicine, 2500 California Plaza, Omaha, NE 68178, USA
| | | |
Collapse
|
29
|
Loss of DAP12 and FcRγ drives exaggerated IL-12 production and CD8(+) T cell response by CCR2(+) Mo-DCs. PLoS One 2013; 8:e76145. [PMID: 24155889 PMCID: PMC3796521 DOI: 10.1371/journal.pone.0076145] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2013] [Accepted: 08/19/2013] [Indexed: 11/24/2022] Open
Abstract
Dap12 and FcRγ, the two transmembrane ITAM-containing signaling adaptors expressed in dendritic cells (DC), are implicated in the regulation of DC function. Several activating and adhesion receptors including integrins require these chains for their function in triggering downstream signaling and effector pathways, however the exact role(s) for Dap12 and FcRγ remains elusive as their loss can lead to both attenuating and enhancing effects. Here, we report that mice congenitally lacking both Dap12 and FcRγ chains (DF) show a massively enhanced effector CD8+ T cell response to protein antigen immunization or West Nile Virus (WNV) infection. Thus, immunization of DF mice with MHCI-restricted OVA peptide leads to accumulation of IL-12-producing monocyte-derived dendritic cells (Mo-DC) in draining lymph nodes, followed by vastly enhanced generation of antigen-specific IFNγ-producing CD8+ T cells. Moreover, DF mice show increased viral clearance in the WNV infection model. Depletion of CCR2+ monocytes/macrophages in vivo by administration anti-CCR2 antibodies or clodronate liposomes completely prevents the exaggerated CD8+ T cell response in DF mice. Mechanistically, we show that the loss of Dap12 and FcRγ-mediated signals in Mo-DC leads to a disruption of GM-CSF receptor-induced STAT5 activation resulting in upregulation of expression of IRF8, a transcription factor. Consequently, Dap12- and FcRγ-deficiency exacerbates GM-CSF-driven monocyte differentiation and production of inflammatory Mo-DC. Our data suggest a novel cross-talk between DC-ITAM and GM-CSF signaling pathways, which controls Mo-DC differentiation, IL-12 production, and CD8+ T cell responses.
Collapse
|
30
|
High levels of FCγR3A and PRF1 expression in peripheral blood mononuclear cells from patients with primary biliary cirrhosis. Dig Dis Sci 2013. [PMID: 23179144 DOI: 10.1007/s10620-012-2456-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
BACKGROUND Innate immunity plays an important role in the pathogenesis of primary biliary cirrhosis (PBC) that needs to be characterized. Levels and clinical relationship of Fc gamma receptor III-A (FcγR3A), tyrosine kinase binding protein (TYROBP) and perforin-1 (PRF1), important genes for nature killer (NK) cells, were analyzed in PBC patients. AIMS The purpose of this study was to explore the expression levels of the above-mentioned genes in peripheral blood mononuclear cells (PBMCs) from PBC patients. METHODS A total of 102 PBC patients and 85 healthy controls (HC) were recruited. The relative levels of FCγR3A, TYROBP, and PRF1 mRNA transcripts in PBMCs were determined by RT-PCR. The percentages of peripheral blood NK, natural killer T (NKT), FCγR3A(+) or PRF1(+) NK cells and PRF1(+) NKT cells in PBC patients and HC were also characterized by flow cytometry analysis. The potential associations of the percentages of NK and NKT cells with clinical indexes were analyzed. RESULTS The relative levels of FCγR3A, TYROBP, and PRF1 mRNA transcripts and the percentages of PRF1(+) NK and NKT cells in PBC patients were significantly higher than that in HC. Moreover, the percentages of PRF1-expressing NK and NKT cells in PBC patients were negatively associated with the levels of serum gamma-glutamyltransferase (GGT) and Mayo risk scores, and the relative levels of FCγR3A expression in NK cells of PBC patients were positively associated with the levels of serum GGT. CONCLUSIONS FCγR3A and PRF1 may participate in the pathogenesis and progression of PBC.
Collapse
|
31
|
Lee SR, Rutan JA, Monteith AJ, Jones SZ, Kang SA, Krum KN, Kilmon MA, Roques JR, Wagner NJ, Clarke SH, Vilen BJ. Receptor cross-talk spatially restricts p-ERK during TLR4 stimulation of autoreactive B cells. THE JOURNAL OF IMMUNOLOGY 2012; 189:3859-68. [PMID: 22984080 DOI: 10.4049/jimmunol.1200940] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
To maintain tolerance, autoreactive B cells must regulate signal transduction from the BCR and TLRs. We recently identified that dendritic cells and macrophages regulate autoreactive cells during TLR4 activation by releasing IL-6 and soluble CD40 ligand (sCD40L). These cytokines selectively repress Ab secretion from autoreactive, but not antigenically naive, B cells. How IL-6 and sCD40L repress autoantibody production is unknown. In this work, we show that IL-6 and sCD40L are required for low-affinity/avidity autoreactive B cells to maintain tolerance through a mechanism involving receptor cross-talk between the BCR, TLR4, and the IL-6R or CD40. We show that acute signaling through IL-6R or CD40 integrates with chronic BCR-mediated ERK activation to restrict p-ERK from the nucleus and represses TLR4-induced Blimp-1 and XBP-1 expression. Tolerance is disrupted in 2-12H/MRL/lpr mice where IL-6 and sCD40L fail to spatially restrict p-ERK and fail to repress TLR4-induced Ig secretion. In the case of CD40, acute signaling in B cells from 2-12H/MRL/lpr mice is intact, but the chronic activation of p-ERK emanating from the BCR is attenuated. Re-establishing chronically active ERK through retroviral expression of constitutively active MEK1 restores tolerance upon sCD40L, but not IL-6, stimulation, indicating that regulation by IL-6 requires another signaling effector. These data define the molecular basis for the regulation of low-affinity autoreactive B cells during TLR4 stimulation; they explain how autoreactive but not naive B cells are repressed by IL-6 and sCD40L; and they identify B cell defects in lupus-prone mice that lead to TLR4-induced autoantibody production.
Collapse
Affiliation(s)
- Sang-Ryul Lee
- Department of Microbiology and Immunology, University of North Carolina, Chapel Hill, NC 27599, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Schafer DP, Lehrman EK, Stevens B. The "quad-partite" synapse: microglia-synapse interactions in the developing and mature CNS. Glia 2012; 61:24-36. [PMID: 22829357 DOI: 10.1002/glia.22389] [Citation(s) in RCA: 406] [Impact Index Per Article: 31.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2012] [Accepted: 06/21/2012] [Indexed: 01/04/2023]
Abstract
Microglia are the resident immune cells and phagocytes of our central nervous system (CNS). While most work has focused on the rapid and robust responses of microglia during CNS disease and injury, emerging evidence suggests that these mysterious cells have important roles at CNS synapses in the healthy, intact CNS. Groundbreaking live imaging studies in the anesthetized, adult mouse demonstrated that microglia processes dynamically survey their environment and interact with other brain cells including neurons and astrocytes. More recent imaging studies have revealed that microglia dynamically interact with synapses where they appear to serve as "synaptic sensors," responding to changes in neural activity and neurotransmitter release. In the following review, we discuss the most recent work demonstrating that microglia play active roles at developing and mature synapses. We first discuss the important imaging studies that have led us to better understand the physical relationship between microglia and synapses in the healthy brain. Following this discussion, we review known molecular mechanisms and functional consequences of microglia-synapse interactions in the developing and mature CNS. Our current knowledge sheds new light on the critical functions of these mysterious cells in synapse development and function in the healthy CNS, but has also incited several new and interesting questions that remain to be explored. We discuss these open questions, and how the most recent findings in the healthy CNS may be related to pathologies associated with abnormal and/or loss of neural circuits.
Collapse
Affiliation(s)
- Dorothy P Schafer
- Department of Neurology, F.M. Kirby Neurobiology Center, Children's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | | | | |
Collapse
|
33
|
Yamanishi Y, Takahashi M, Izawa K, Isobe M, Ito S, Tsuchiya A, Maehara A, Kaitani A, Uchida T, Togami K, Enomoto Y, Nakahara F, Oki T, Kajikawa M, Kurihara H, Kitamura T, Kitaura J. A Soluble Form of LMIR5/CD300b Amplifies Lipopolysaccharide-Induced Lethal Inflammation in Sepsis. THE JOURNAL OF IMMUNOLOGY 2012; 189:1773-9. [DOI: 10.4049/jimmunol.1201139] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
|
34
|
Huynh L, Wang L, Shi C, Park-Min KH, Ivashkiv LB. ITAM-coupled receptors inhibit IFNAR signaling and alter macrophage responses to TLR4 and Listeria monocytogenes. THE JOURNAL OF IMMUNOLOGY 2012; 188:3447-57. [PMID: 22368279 DOI: 10.4049/jimmunol.1102211] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
ITAM-coupled receptors play an essential role in regulating macrophage activation and function by cross-regulating signaling from heterologous receptors. We investigated mechanisms by which ITAM-associated receptors inhibit type I IFN (IFN-α/β) signaling in primary human macrophages and tested the effects of simultaneous ligation of ITAM-associated receptors and TLR4 on TLR4-induced Jak-STAT signaling that is mediated by autocrine IFN-β. Preligation of ITAM-coupled β2 integrins and FcγRs inhibited proximal signaling by the type I IFN receptor IFNAR. Cross-inhibition of IFNAR signaling by β2 integrins resulted in decreased Jak1 activation and was mediated by partial downregulation of the IFNAR1 subunit and MAPK-dependent induction of USP18, which blocks the association of Jak1 with IFNAR2. Simultaneous engagement of ITAM-coupled β2 integrins or Dectin-1 with TLR4 did not affect TLR4-induced direct activation of inflammatory target genes such as TNF or IL6 but abrogated subsequent induction of IFN response genes that is mediated by autocrine IFN-β signaling. Type I IFNs promote macrophage death postinfection by Listeria monocytogenes. Consequently, attenuation of IFN responses by β2 integrins protected primary human macrophages from L. monocytogenes-induced apoptosis. These results provide a mechanism for cross-inhibition of type I IFN signaling by ITAM-coupled β2 integrins and demonstrate that ITAM signaling qualitatively modulates macrophage responses to pathogen-associated molecular patterns and pathogens by selectively suppressing IFN responses.
Collapse
Affiliation(s)
- Linda Huynh
- Graduate Program in Immunology and Microbial Pathogenesis, Weill Cornell Graduate School of Medical Sciences, New York, NY 10021, USA
| | | | | | | | | |
Collapse
|
35
|
Zhu M, Fuller DM, Ou-Yang CW, Sullivan SA, Zhang W. Tyrosine phosphorylation-independent regulation of lipopolysaccharide-mediated response by the transmembrane adaptor protein LAB. THE JOURNAL OF IMMUNOLOGY 2012; 188:2733-41. [PMID: 22308309 DOI: 10.4049/jimmunol.1101581] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Linker for activation of B cells (LAB)/non-T cell activation linker is a transmembrane adaptor protein that functions in immunoreceptor-mediated signaling. Published studies have shown that LAB has both positive and negative roles in regulating TCR and high-affinity Fc receptor-mediated signaling and cellular function. In this study, we showed that LAB was also expressed in dendritic cells and that LAB deficiency affected LPS-mediated signaling and cytokine production. LPS-mediated MAPK activation was enhanced in LAB(-/-) bone marrow-derived dendritic cells. These bone marrow-derived dendritic cells also produced more TNF-α, IL-6, and IL-10 than wild-type cells. Moreover, LAB(-/-) mice were hyperresponsive to LPS-induced septic shock. These data indicated that LAB has a negative role in LPS-mediated responses. By using LAB knockin mice, which harbor mutations at five membrane-distal tyrosines, we further showed that, in contrast to its role in immunoreceptor-mediated signaling, LAB function in LPS-mediated signaling pathway did not depend on its tyrosine phosphorylation. Our study suggested a novel mechanism by which LAB functions in the regulation of innate immunity.
Collapse
Affiliation(s)
- Minghua Zhu
- Department of Immunology, Duke University Medical Center, Durham, NC 27710, USA
| | | | | | | | | |
Collapse
|
36
|
The prolyl isomerase Pin1 modulates development of CD8+ cDC in mice. PLoS One 2012; 7:e29808. [PMID: 22238658 PMCID: PMC3251613 DOI: 10.1371/journal.pone.0029808] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2011] [Accepted: 12/06/2011] [Indexed: 11/21/2022] Open
Abstract
Background Pin1 has previously been described to regulate cells that participate in both innate and adaptive immunity. Thus far, however, no role for Pin1 has been described in modulating conventional dendritic cells, innate antigen presenting cells that potently activate naïve T cells, thereby bridging innate and adaptive immune responses. Methodology/Principal Findings When challenged with LPS, Pin1-null mice failed to accumulate spleen conventional dendritic cells (cDC). Analysis of steady-state spleen DC populations revealed that Pin1-null mice had fewer CD8+ cDC. This defect was recapitulated by culturing Pin1-null bone marrow with the DC-instructive cytokine Flt3 Ligand. Additionally, injection of Flt3 Ligand for 9 days failed to induce robust expansion of CD8+ cDC in Pin1-null mice. Upon infection with Listeria monocytogenes, Pin1-null mice were defective in stimulating proliferation of adoptively transferred WT CD8+ T cells, suggesting that decreases in Pin1 null CD8+ cDC may affect T cell responses to infection in vivo. Finally, upon analyzing expression of proteins involved in DC development, elevated expression of PU.1 was detected in Pin1-null cells, which resulted from an increase in PU.1 protein half-life. Conclusions/Significance We have identified a novel role for Pin1 as a modulator of CD8+ cDC development. Consistent with reduced numbers of CD8+ cDC in Pin1-null mice, we find that the absence of Pin1 impairs CD8+ T cell proliferation in response to infection with Listeria monocytogenes. These data suggest that, via regulation of CD8+ cDC production, Pin1 may serve as an important modulator of adaptive immunity.
Collapse
|
37
|
Srivastava RM, Srivastava S, Singh M, Bajpai VK, Ghosh JK. Consequences of alteration in leucine zipper sequence of melittin in its neutralization of lipopolysaccharide-induced proinflammatory response in macrophage cells and interaction with lipopolysaccharide. J Biol Chem 2011; 287:1980-95. [PMID: 22128186 DOI: 10.1074/jbc.m111.302893] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The bee venom antimicrobial peptide, melittin, besides showing versatile activity against microorganisms also neutralizes lipopolysaccharide (LPS)-induced proinflammatory responses in macrophage cells. However, how the amino acid sequence of melittin contributes in its anti-inflammatory properties is mostly unknown. To determine the importance of the leucine zipper sequence of melittin in its neutralization of LPS-induced inflammatory responses in macrophages and interaction with LPS, anti-inflammatory properties of melittin and its three analogues and their interactions with LPS were studied in detail. Two of these analogues, namely melittin Mut-1 (MM-1) and melittin Mut-2 (MM-2), possess leucine to alanine substitutions in the single and double heptadic leucine residue(s) of melittin, respectively, whereas the third analogue is a scrambled peptide (Mel-SCR) that contains the amino acid composition of melittin with minor rearrangement in its leucine zipper sequence. Although MM-1 partly inhibited the production of proinflammatory cytokines in RAW 264.7 and rat primary macrophage cells in the presence of LPS, MM-2 and Mel-SCR were negligibly active. A progressive decrease in interaction of melittin with LPS, aggregation in LPS, and dissociation of LPS aggregates with alteration in the leucine zipper sequence of melittin was observed. Furthermore, with alteration in the leucine zipper sequence of melittin, these analogues failed to exhibit cellular responses associated with neutralization of LPS-induced inflammatory responses in macrophage cells by melittin. The data indicated a probable important role of the leucine zipper sequence of melittin in neutralizing LPS-induced proinflammatory responses in macrophage cells as well as in its interaction with LPS.
Collapse
Affiliation(s)
- Raghvendra M Srivastava
- Molecular and Structural Biology Division, Sophisticated Analytical Instrument Facility, Central Drug Research Institute, Council of Scientific and Industrial Research, Chattar Manzil Palace, P. O. Box 173, Lucknow 226001, India
| | | | | | | | | |
Collapse
|
38
|
Involvement of the TREM-1/DAP12 pathway in the innate immune responses to Porphyromonas gingivalis. Mol Immunol 2011; 49:387-94. [DOI: 10.1016/j.molimm.2011.09.012] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2011] [Revised: 09/15/2011] [Accepted: 09/16/2011] [Indexed: 11/17/2022]
|
39
|
McCormick S, Shaler CR, Small CL, Horvath C, Damjanovic D, Brown EG, Aoki N, Takai T, Xing Z. Control of pathogenic CD4 T cells and lethal immunopathology by signaling immunoadaptor DAP12 during influenza infection. THE JOURNAL OF IMMUNOLOGY 2011; 187:4280-92. [PMID: 21908731 DOI: 10.4049/jimmunol.1101050] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Immunopathology is a major cause of influenza-associated morbidity and mortality worldwide. However, the role and regulatory mechanisms of CD4 T cells in severe lung immunopathology following acute influenza infection are poorly understood. In this paper, we report that the emergence of immunopathogenic CD4 T cells is under the control of a transmembrane immunoadaptor DAP12 pathway during influenza infection. We find that the mice lacking DAP12 have unaltered viral clearance but easily succumb to influenza infection as a result of uncontrolled immunopathology. Such immunopathology is associated with markedly increased CD4 T cells displaying markedly increased cytotoxicity and Fas ligand expression. Furthermore, the immunopathogenic property of these CD4 T cells is transferrable. Thus, depletion of CD4 T cells or abrogation of Fas/Fas ligand signaling pathway improves survival and immunopathology. We further find that DAP12 expressed by dendritic cells plays an important role in controlling the immunopathogenic CD4 T cells during influenza infection. Our findings identify a novel pathway that controls the level of immune-pathogenic CD4 T cells during acute influenza infection.
Collapse
Affiliation(s)
- Sarah McCormick
- Department of Pathology and Molecular Medicine, McMaster Immunology Research Centre, McMaster University, Hamilton, Ontario L8S 4K1, Canada
| | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Barrow AD, Raynal N, Andersen TL, Slatter DA, Bihan D, Pugh N, Cella M, Kim T, Rho J, Negishi-Koga T, Delaisse JM, Takayanagi H, Lorenzo J, Colonna M, Farndale RW, Choi Y, Trowsdale J. OSCAR is a collagen receptor that costimulates osteoclastogenesis in DAP12-deficient humans and mice. J Clin Invest 2011; 121:3505-16. [PMID: 21841309 PMCID: PMC3163954 DOI: 10.1172/jci45913] [Citation(s) in RCA: 152] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2010] [Accepted: 07/01/2011] [Indexed: 12/14/2022] Open
Abstract
Osteoclasts are terminally differentiated leukocytes that erode the mineralized bone matrix. Osteoclastogenesis requires costimulatory receptor signaling through adaptors containing immunoreceptor tyrosine-based activation motifs (ITAMs), such as Fc receptor common γ (FcRγ) and DNAX-activating protein of 12 kDa. Identification of these ITAM-containing receptors and their ligands remains a high research priority, since the stimuli for osteoclastogenesis are only partly defined. Osteoclast-associated receptor (OSCAR) was proposed to be a potent FcRγ-associated costimulatory receptor expressed by preosteoclasts in vitro, but OSCAR lacks a cognate ligand and its role in vivo has been unclear. Using samples from mice and patients deficient in various ITAM signaling pathways, we show here that OSCAR costimulates one of the major FcRγ-associated pathways required for osteoclastogenesis in vivo. Furthermore, we found that OSCAR binds to specific motifs within fibrillar collagens in the ECM that become revealed on nonquiescent bone surfaces in which osteoclasts undergo maturation and terminal differentiation in vivo. OSCAR promoted osteoclastogenesis in vivo, and OSCAR binding to its collagen motif led to signaling that increased numbers of osteoclasts in culture. Thus, our results suggest that ITAM-containing receptors can respond to exposed ligands in collagen, leading to the functional differentiation of leukocytes, which provides what we believe to be a new concept for ITAM regulation of cytokine receptors in different tissue microenvironments.
Collapse
Affiliation(s)
- Alexander David Barrow
- Department of Pathology, University of Cambridge, Cambridge, United Kingdom.
Washington University School of Medicine, Department of Pathology and Immunology, St. Louis, Missouri, USA.
Department of Biochemistry, University of Cambridge, Cambridge, United Kingdom.
Department of Clinical Cell Biology, University of Southern Denmark, Vejle/Lillebælt Hospital, Institute of Regional Health Services Research, Vejle, Denmark.
Department of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA.
Department of Microbiology, BK21 BioBC, and Graduate of Analytical Science and Technology, Chungnam National University, Yuseong-gu, Daejon, Republic of Korea.
Department of Cell Signaling, Tokyo Medical and Dental University, Tokyo, Japan.
Division of Endocrinology, Department of Medicine, University of Connecticut Health Center, Farmington, Connecticut, USA
| | - Nicolas Raynal
- Department of Pathology, University of Cambridge, Cambridge, United Kingdom.
Washington University School of Medicine, Department of Pathology and Immunology, St. Louis, Missouri, USA.
Department of Biochemistry, University of Cambridge, Cambridge, United Kingdom.
Department of Clinical Cell Biology, University of Southern Denmark, Vejle/Lillebælt Hospital, Institute of Regional Health Services Research, Vejle, Denmark.
Department of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA.
Department of Microbiology, BK21 BioBC, and Graduate of Analytical Science and Technology, Chungnam National University, Yuseong-gu, Daejon, Republic of Korea.
Department of Cell Signaling, Tokyo Medical and Dental University, Tokyo, Japan.
Division of Endocrinology, Department of Medicine, University of Connecticut Health Center, Farmington, Connecticut, USA
| | - Thomas Levin Andersen
- Department of Pathology, University of Cambridge, Cambridge, United Kingdom.
Washington University School of Medicine, Department of Pathology and Immunology, St. Louis, Missouri, USA.
Department of Biochemistry, University of Cambridge, Cambridge, United Kingdom.
Department of Clinical Cell Biology, University of Southern Denmark, Vejle/Lillebælt Hospital, Institute of Regional Health Services Research, Vejle, Denmark.
Department of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA.
Department of Microbiology, BK21 BioBC, and Graduate of Analytical Science and Technology, Chungnam National University, Yuseong-gu, Daejon, Republic of Korea.
Department of Cell Signaling, Tokyo Medical and Dental University, Tokyo, Japan.
Division of Endocrinology, Department of Medicine, University of Connecticut Health Center, Farmington, Connecticut, USA
| | - David A. Slatter
- Department of Pathology, University of Cambridge, Cambridge, United Kingdom.
Washington University School of Medicine, Department of Pathology and Immunology, St. Louis, Missouri, USA.
Department of Biochemistry, University of Cambridge, Cambridge, United Kingdom.
Department of Clinical Cell Biology, University of Southern Denmark, Vejle/Lillebælt Hospital, Institute of Regional Health Services Research, Vejle, Denmark.
Department of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA.
Department of Microbiology, BK21 BioBC, and Graduate of Analytical Science and Technology, Chungnam National University, Yuseong-gu, Daejon, Republic of Korea.
Department of Cell Signaling, Tokyo Medical and Dental University, Tokyo, Japan.
Division of Endocrinology, Department of Medicine, University of Connecticut Health Center, Farmington, Connecticut, USA
| | - Dominique Bihan
- Department of Pathology, University of Cambridge, Cambridge, United Kingdom.
Washington University School of Medicine, Department of Pathology and Immunology, St. Louis, Missouri, USA.
Department of Biochemistry, University of Cambridge, Cambridge, United Kingdom.
Department of Clinical Cell Biology, University of Southern Denmark, Vejle/Lillebælt Hospital, Institute of Regional Health Services Research, Vejle, Denmark.
Department of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA.
Department of Microbiology, BK21 BioBC, and Graduate of Analytical Science and Technology, Chungnam National University, Yuseong-gu, Daejon, Republic of Korea.
Department of Cell Signaling, Tokyo Medical and Dental University, Tokyo, Japan.
Division of Endocrinology, Department of Medicine, University of Connecticut Health Center, Farmington, Connecticut, USA
| | - Nicholas Pugh
- Department of Pathology, University of Cambridge, Cambridge, United Kingdom.
Washington University School of Medicine, Department of Pathology and Immunology, St. Louis, Missouri, USA.
Department of Biochemistry, University of Cambridge, Cambridge, United Kingdom.
Department of Clinical Cell Biology, University of Southern Denmark, Vejle/Lillebælt Hospital, Institute of Regional Health Services Research, Vejle, Denmark.
Department of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA.
Department of Microbiology, BK21 BioBC, and Graduate of Analytical Science and Technology, Chungnam National University, Yuseong-gu, Daejon, Republic of Korea.
Department of Cell Signaling, Tokyo Medical and Dental University, Tokyo, Japan.
Division of Endocrinology, Department of Medicine, University of Connecticut Health Center, Farmington, Connecticut, USA
| | - Marina Cella
- Department of Pathology, University of Cambridge, Cambridge, United Kingdom.
Washington University School of Medicine, Department of Pathology and Immunology, St. Louis, Missouri, USA.
Department of Biochemistry, University of Cambridge, Cambridge, United Kingdom.
Department of Clinical Cell Biology, University of Southern Denmark, Vejle/Lillebælt Hospital, Institute of Regional Health Services Research, Vejle, Denmark.
Department of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA.
Department of Microbiology, BK21 BioBC, and Graduate of Analytical Science and Technology, Chungnam National University, Yuseong-gu, Daejon, Republic of Korea.
Department of Cell Signaling, Tokyo Medical and Dental University, Tokyo, Japan.
Division of Endocrinology, Department of Medicine, University of Connecticut Health Center, Farmington, Connecticut, USA
| | - Taesoo Kim
- Department of Pathology, University of Cambridge, Cambridge, United Kingdom.
Washington University School of Medicine, Department of Pathology and Immunology, St. Louis, Missouri, USA.
Department of Biochemistry, University of Cambridge, Cambridge, United Kingdom.
Department of Clinical Cell Biology, University of Southern Denmark, Vejle/Lillebælt Hospital, Institute of Regional Health Services Research, Vejle, Denmark.
Department of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA.
Department of Microbiology, BK21 BioBC, and Graduate of Analytical Science and Technology, Chungnam National University, Yuseong-gu, Daejon, Republic of Korea.
Department of Cell Signaling, Tokyo Medical and Dental University, Tokyo, Japan.
Division of Endocrinology, Department of Medicine, University of Connecticut Health Center, Farmington, Connecticut, USA
| | - Jaerang Rho
- Department of Pathology, University of Cambridge, Cambridge, United Kingdom.
Washington University School of Medicine, Department of Pathology and Immunology, St. Louis, Missouri, USA.
Department of Biochemistry, University of Cambridge, Cambridge, United Kingdom.
Department of Clinical Cell Biology, University of Southern Denmark, Vejle/Lillebælt Hospital, Institute of Regional Health Services Research, Vejle, Denmark.
Department of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA.
Department of Microbiology, BK21 BioBC, and Graduate of Analytical Science and Technology, Chungnam National University, Yuseong-gu, Daejon, Republic of Korea.
Department of Cell Signaling, Tokyo Medical and Dental University, Tokyo, Japan.
Division of Endocrinology, Department of Medicine, University of Connecticut Health Center, Farmington, Connecticut, USA
| | - Takako Negishi-Koga
- Department of Pathology, University of Cambridge, Cambridge, United Kingdom.
Washington University School of Medicine, Department of Pathology and Immunology, St. Louis, Missouri, USA.
Department of Biochemistry, University of Cambridge, Cambridge, United Kingdom.
Department of Clinical Cell Biology, University of Southern Denmark, Vejle/Lillebælt Hospital, Institute of Regional Health Services Research, Vejle, Denmark.
Department of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA.
Department of Microbiology, BK21 BioBC, and Graduate of Analytical Science and Technology, Chungnam National University, Yuseong-gu, Daejon, Republic of Korea.
Department of Cell Signaling, Tokyo Medical and Dental University, Tokyo, Japan.
Division of Endocrinology, Department of Medicine, University of Connecticut Health Center, Farmington, Connecticut, USA
| | - Jean-Marie Delaisse
- Department of Pathology, University of Cambridge, Cambridge, United Kingdom.
Washington University School of Medicine, Department of Pathology and Immunology, St. Louis, Missouri, USA.
Department of Biochemistry, University of Cambridge, Cambridge, United Kingdom.
Department of Clinical Cell Biology, University of Southern Denmark, Vejle/Lillebælt Hospital, Institute of Regional Health Services Research, Vejle, Denmark.
Department of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA.
Department of Microbiology, BK21 BioBC, and Graduate of Analytical Science and Technology, Chungnam National University, Yuseong-gu, Daejon, Republic of Korea.
Department of Cell Signaling, Tokyo Medical and Dental University, Tokyo, Japan.
Division of Endocrinology, Department of Medicine, University of Connecticut Health Center, Farmington, Connecticut, USA
| | - Hiroshi Takayanagi
- Department of Pathology, University of Cambridge, Cambridge, United Kingdom.
Washington University School of Medicine, Department of Pathology and Immunology, St. Louis, Missouri, USA.
Department of Biochemistry, University of Cambridge, Cambridge, United Kingdom.
Department of Clinical Cell Biology, University of Southern Denmark, Vejle/Lillebælt Hospital, Institute of Regional Health Services Research, Vejle, Denmark.
Department of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA.
Department of Microbiology, BK21 BioBC, and Graduate of Analytical Science and Technology, Chungnam National University, Yuseong-gu, Daejon, Republic of Korea.
Department of Cell Signaling, Tokyo Medical and Dental University, Tokyo, Japan.
Division of Endocrinology, Department of Medicine, University of Connecticut Health Center, Farmington, Connecticut, USA
| | - Joseph Lorenzo
- Department of Pathology, University of Cambridge, Cambridge, United Kingdom.
Washington University School of Medicine, Department of Pathology and Immunology, St. Louis, Missouri, USA.
Department of Biochemistry, University of Cambridge, Cambridge, United Kingdom.
Department of Clinical Cell Biology, University of Southern Denmark, Vejle/Lillebælt Hospital, Institute of Regional Health Services Research, Vejle, Denmark.
Department of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA.
Department of Microbiology, BK21 BioBC, and Graduate of Analytical Science and Technology, Chungnam National University, Yuseong-gu, Daejon, Republic of Korea.
Department of Cell Signaling, Tokyo Medical and Dental University, Tokyo, Japan.
Division of Endocrinology, Department of Medicine, University of Connecticut Health Center, Farmington, Connecticut, USA
| | - Marco Colonna
- Department of Pathology, University of Cambridge, Cambridge, United Kingdom.
Washington University School of Medicine, Department of Pathology and Immunology, St. Louis, Missouri, USA.
Department of Biochemistry, University of Cambridge, Cambridge, United Kingdom.
Department of Clinical Cell Biology, University of Southern Denmark, Vejle/Lillebælt Hospital, Institute of Regional Health Services Research, Vejle, Denmark.
Department of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA.
Department of Microbiology, BK21 BioBC, and Graduate of Analytical Science and Technology, Chungnam National University, Yuseong-gu, Daejon, Republic of Korea.
Department of Cell Signaling, Tokyo Medical and Dental University, Tokyo, Japan.
Division of Endocrinology, Department of Medicine, University of Connecticut Health Center, Farmington, Connecticut, USA
| | - Richard W. Farndale
- Department of Pathology, University of Cambridge, Cambridge, United Kingdom.
Washington University School of Medicine, Department of Pathology and Immunology, St. Louis, Missouri, USA.
Department of Biochemistry, University of Cambridge, Cambridge, United Kingdom.
Department of Clinical Cell Biology, University of Southern Denmark, Vejle/Lillebælt Hospital, Institute of Regional Health Services Research, Vejle, Denmark.
Department of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA.
Department of Microbiology, BK21 BioBC, and Graduate of Analytical Science and Technology, Chungnam National University, Yuseong-gu, Daejon, Republic of Korea.
Department of Cell Signaling, Tokyo Medical and Dental University, Tokyo, Japan.
Division of Endocrinology, Department of Medicine, University of Connecticut Health Center, Farmington, Connecticut, USA
| | - Yongwon Choi
- Department of Pathology, University of Cambridge, Cambridge, United Kingdom.
Washington University School of Medicine, Department of Pathology and Immunology, St. Louis, Missouri, USA.
Department of Biochemistry, University of Cambridge, Cambridge, United Kingdom.
Department of Clinical Cell Biology, University of Southern Denmark, Vejle/Lillebælt Hospital, Institute of Regional Health Services Research, Vejle, Denmark.
Department of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA.
Department of Microbiology, BK21 BioBC, and Graduate of Analytical Science and Technology, Chungnam National University, Yuseong-gu, Daejon, Republic of Korea.
Department of Cell Signaling, Tokyo Medical and Dental University, Tokyo, Japan.
Division of Endocrinology, Department of Medicine, University of Connecticut Health Center, Farmington, Connecticut, USA
| | - John Trowsdale
- Department of Pathology, University of Cambridge, Cambridge, United Kingdom.
Washington University School of Medicine, Department of Pathology and Immunology, St. Louis, Missouri, USA.
Department of Biochemistry, University of Cambridge, Cambridge, United Kingdom.
Department of Clinical Cell Biology, University of Southern Denmark, Vejle/Lillebælt Hospital, Institute of Regional Health Services Research, Vejle, Denmark.
Department of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA.
Department of Microbiology, BK21 BioBC, and Graduate of Analytical Science and Technology, Chungnam National University, Yuseong-gu, Daejon, Republic of Korea.
Department of Cell Signaling, Tokyo Medical and Dental University, Tokyo, Japan.
Division of Endocrinology, Department of Medicine, University of Connecticut Health Center, Farmington, Connecticut, USA
| |
Collapse
|
41
|
Sumpter TL, Packiam V, Turnquist HR, Castellaneta A, Yoshida O, Thomson AW. DAP12 promotes IRAK-M expression and IL-10 production by liver myeloid dendritic cells and restrains their T cell allostimulatory ability. THE JOURNAL OF IMMUNOLOGY 2011; 186:1970-80. [PMID: 21257958 DOI: 10.4049/jimmunol.1000527] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Freshly isolated hepatic dendritic cells (DC) are comparatively immature, relatively resistant to maturation, and can downmodulate effector T cell responses. Molecular mechanisms that underlie these properties are ill defined. DNAX-activating protein of 12 kDa (DAP12) is an ITAM-bearing transmembrane adaptor protein that integrates signals through several receptors, including triggering receptor expressed on myeloid cells-1, -2, and CD200R. Notably, DC propagated from DAP12-deficient mice exhibit enhanced maturation in response to TLR ligation. Given the constitutive exposure of liver DC to endotoxin draining from the gut, we hypothesized that DAP12 might regulate liver DC maturation. We show that DAP12 is expressed by freshly isolated liver, spleen, kidney, and lung myeloid DC. Moreover, inhibition of DAP12 expression by liver DC using small interfering RNA promotes their phenotypic and functional maturation, resulting in enhanced TNF-α, IL-6, and IL-12p70 production, reduced secretion of IL-10, and enhanced CD4(+) and CD8(+) T cell proliferation. Furthermore, DAP12 silencing correlates with decreased STAT3 phosphorylation in mature liver DC and with diminished expression of the IL-1R-associated kinase-M, a negative regulator of TLR signaling. These findings highlight a regulatory role for DAP12 in hepatic DC maturation, and suggest a mechanism whereby this function may be induced/maintained.
Collapse
Affiliation(s)
- Tina L Sumpter
- Starzl Transplantation Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | | | | | | | | | | |
Collapse
|
42
|
Smrz D, Iwaki S, McVicar DW, Metcalfe DD, Gilfillan AM. TLR-mediated signaling pathways circumvent the requirement for DAP12 in mast cells for the induction of inflammatory mediator release. Eur J Immunol 2010; 40:3557-69. [PMID: 21108475 DOI: 10.1002/eji.201040573] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2010] [Revised: 08/02/2010] [Accepted: 09/13/2010] [Indexed: 02/06/2023]
Abstract
TLR, expressed on the surface of mast cells, respond to a variety of bacterial and viral components to induce and enhance high-affinity IgE receptor-mediated cytokine production. Recent reports have indicated that specific TLR-dependent responses in macrophages and dendritic cells are regulated by the ITAM-containing molecule, DAP12. When phosphorylated, DAP12 recruits Syk, which is a critical molecule for mast cell activation. We therefore examined whether DAP12 similarly regulates TLR-mediated responses in mast cells. DAP12 was confirmed to be expressed in both human and mouse mast cells and, upon phosphorylation, to recruit Syk. However, although TLR agonists induced cytokine production, and synergistically enhanced high-affinity IgE receptor-mediated cytokine production, surprisingly, they failed to increase DAP12 phosphorylation in mouse bone marrow-derived mast cells (BMMC). Furthermore, normal TLR-mediated responses were observed in DAP12(-/-) BMMC. However, DAP12 phosphorylation and subsequent Syk recruitment were observed in BMMC following Con A-induced aggregation of mannose-glycosylated receptors, and these responses, together with Con A-induced degranulation, were substantially reduced in the DAP12(-/-) BMMC. These data demonstrate that TLR have differential requirements for DAP12 for their function in different cell types and that the inability of TLR to influence mast cell degranulation may be linked to their inability to utilize DAP12 to recruit Syk.
Collapse
Affiliation(s)
- Daniel Smrz
- Laboratory of Allergic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892-1881, USA
| | | | | | | | | |
Collapse
|
43
|
Fernandez PL, Dutra FF, Alves L, Figueiredo RT, Mourão-Sa D, Fortes GB, Bergstrand S, Lönn D, Cevallos RR, Pereira RMS, Lopes UG, Travassos LH, Paiva CN, Bozza MT. Heme amplifies the innate immune response to microbial molecules through spleen tyrosine kinase (Syk)-dependent reactive oxygen species generation. J Biol Chem 2010; 285:32844-32851. [PMID: 20729208 DOI: 10.1074/jbc.m110.146076] [Citation(s) in RCA: 73] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Infectious diseases that cause hemolysis are among the most threatening human diseases, because of severity and/or global distribution. In these conditions, hemeproteins and heme are released, but whether heme affects the inflammatory response to microorganism molecules remains to be characterized. Here, we show that heme increased the lethality and cytokine secretion induced by LPS in vivo and enhanced the secretion of cytokines by macrophages stimulated with various agonists of innate immune receptors. Activation of nuclear factor κB (NF-κB) and MAPKs and the generation of reactive oxygen species were essential to the increase in cytokine production induced by heme plus LPS. This synergistic effect of heme and LPS was blocked by a selective inhibitor of spleen tyrosine kinase (Syk) and was abrogated in dendritic cells deficient in Syk. Moreover, inhibition of Syk and the downstream molecules PKC and PI3K reduced the reactive oxygen species generation by heme. Our results highlight a mechanism by which heme amplifies the secretion of cytokines triggered by microbial molecule activation and indicates possible pathways for therapeutic intervention during hemolytic infectious diseases.
Collapse
Affiliation(s)
- Patricia L Fernandez
- From the Laboratório de Inflamação e Imunidade, Departamento de Imunologia, Instituto de Microbiologia, Universidade Federal do Rio de Janeiro, 21941-902 Rio de Janeiro, Brazil; Instituto de Investigaciones Científicas y Servicios de Alta Tecnologia, 0843-01103 Ciudad de Panamá, Panamá
| | - Fabianno F Dutra
- From the Laboratório de Inflamação e Imunidade, Departamento de Imunologia, Instituto de Microbiologia, Universidade Federal do Rio de Janeiro, 21941-902 Rio de Janeiro, Brazil
| | - Letícia Alves
- From the Laboratório de Inflamação e Imunidade, Departamento de Imunologia, Instituto de Microbiologia, Universidade Federal do Rio de Janeiro, 21941-902 Rio de Janeiro, Brazil
| | - Rodrigo T Figueiredo
- From the Laboratório de Inflamação e Imunidade, Departamento de Imunologia, Instituto de Microbiologia, Universidade Federal do Rio de Janeiro, 21941-902 Rio de Janeiro, Brazil; Pólo de Xerém, Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, 21941-902 Rio de Janeiro, Brazil
| | - Diego Mourão-Sa
- Immunobiology Laboratory, Cancer Research UK, London Research Institute, Lincoln's Inn Fields Laboratories, London WC2A 3PX, United Kingdom
| | - Guilherme B Fortes
- From the Laboratório de Inflamação e Imunidade, Departamento de Imunologia, Instituto de Microbiologia, Universidade Federal do Rio de Janeiro, 21941-902 Rio de Janeiro, Brazil
| | - Sophie Bergstrand
- From the Laboratório de Inflamação e Imunidade, Departamento de Imunologia, Instituto de Microbiologia, Universidade Federal do Rio de Janeiro, 21941-902 Rio de Janeiro, Brazil
| | - David Lönn
- From the Laboratório de Inflamação e Imunidade, Departamento de Imunologia, Instituto de Microbiologia, Universidade Federal do Rio de Janeiro, 21941-902 Rio de Janeiro, Brazil
| | - Ricardo R Cevallos
- Instituto de Investigaciones Científicas y Servicios de Alta Tecnologia, 0843-01103 Ciudad de Panamá, Panamá
| | - Renata M S Pereira
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, 21941-902 Rio de Janeiro, Brazil
| | - Ulisses G Lopes
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, 21941-902 Rio de Janeiro, Brazil
| | - Leonardo H Travassos
- From the Laboratório de Inflamação e Imunidade, Departamento de Imunologia, Instituto de Microbiologia, Universidade Federal do Rio de Janeiro, 21941-902 Rio de Janeiro, Brazil; Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, 21941-902 Rio de Janeiro, Brazil
| | - Claudia N Paiva
- From the Laboratório de Inflamação e Imunidade, Departamento de Imunologia, Instituto de Microbiologia, Universidade Federal do Rio de Janeiro, 21941-902 Rio de Janeiro, Brazil
| | - Marcelo T Bozza
- From the Laboratório de Inflamação e Imunidade, Departamento de Imunologia, Instituto de Microbiologia, Universidade Federal do Rio de Janeiro, 21941-902 Rio de Janeiro, Brazil.
| |
Collapse
|
44
|
ITAM signaling in dendritic cells controls T helper cell priming by regulating MHC class II recycling. Blood 2010; 116:3208-18. [PMID: 20634378 DOI: 10.1182/blood-2009-10-250415] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Immature dendritic cells (DCs) specialize in antigen capture and maintain a highly dynamic pool of intracellular major histocompatibility complex class II (MHCII) that continuously recycles from peptide loading compartments to the plasma membrane and back again. This process facilitates sampling of environmental antigens for presentation to T helper cells. Here, we show that a signaling pathway mediated by the DC immunoreceptor tyrosine-based activation motif (ITAM)-containing adaptors (DAP12 and FcRγ) and Vav family guanine nucleotide exchange factors controls the half-life of surface peptide-MHCII (pMHCII) complexes and is critical for CD4 T-cell triggering in vitro. Strikingly, mice with disrupted DC ITAMs show defective T helper cell priming in vivo and are protected from experimental autoimmune encephalitis. Mechanistically, we show that deficiency in ITAM signaling results in increased pMHCII internalization, impaired recycling, and an accumulation of ubiquitinated MHCII species that are prematurely degraded in lysosomes. We propose a novel mechanism for control of T helper cell priming.
Collapse
|
45
|
Wang L, Gordon RA, Huynh L, Su X, Park Min KH, Han J, Arthur JS, Kalliolias GD, Ivashkiv LB. Indirect inhibition of Toll-like receptor and type I interferon responses by ITAM-coupled receptors and integrins. Immunity 2010; 32:518-30. [PMID: 20362473 DOI: 10.1016/j.immuni.2010.03.014] [Citation(s) in RCA: 117] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2009] [Revised: 11/25/2009] [Accepted: 03/19/2010] [Indexed: 01/27/2023]
Abstract
An important function of immunoreceptor tyrosine-based activation motif (ITAM)-coupled receptors is cross-regulation of heterologous receptor signaling, but mechanisms of cross-inhibition are poorly understood. We show that high-avidity ligation of ITAM-coupled beta2 integrins and FcgammaRs in macrophages inhibited type I interferon receptor and Toll-like receptor (TLR) signaling and induced expression of interleukin-10 (IL-10); signaling inhibitors SOCS3, ABIN-3, and A20; and repressors of cytokine gene transcription STAT3 and Hes1. Induction of inhibitors was dependent on a pathway composed of signaling molecules DAP12, Syk, and Pyk2 that coupled to downstream kinases p38 and MSKs and required integration of IL-10-dependent and -independent signals. ITAM-induced inhibitors abrogated TLR responses by cooperatively targeting distinct steps in TLR signaling. Inhibitory signaling was suppressed by IFN-gamma and attenuated in inflammatory arthritis synovial macrophages. These results provide an indirect mechanism of cross-inhibition of TLRs and delineate a signaling pathway important for deactivation of macrophages.
Collapse
Affiliation(s)
- Lu Wang
- Arthritis and Tissue Degeneration Program, Hospital for Special Surgery, New York, NY 10021, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Regulation of Toll-like receptor signaling in the innate immunity. SCIENCE CHINA-LIFE SCIENCES 2010; 53:34-43. [PMID: 20596954 DOI: 10.1007/s11427-010-0011-x] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/27/2009] [Accepted: 12/10/2009] [Indexed: 12/12/2022]
Abstract
Toll-like receptors sense invading pathogens by recognizing a wide variety of conserved pathogen-associated molecular patterns (PAMPs). The members of TLR family selectively utilize adaptor proteins MyD88, TRIF, TIRAP and TRAM to activate overlapping but distinct signal transduction pathways which trigger production of different panels of mediators such as proinflammatory cytokines and type I interferon. These mediators not only control innate immunity but also direct subsequently developed adaptive immunity. TLR activation is strictly and finely regulated at multiple levels of the signal transduction pathways.
Collapse
|
47
|
Modulation of paired immunoglobulin-like type 2 receptor signaling alters the host response to Staphylococcus aureus-induced pneumonia. Infect Immun 2010; 78:1353-63. [PMID: 20065029 DOI: 10.1128/iai.00969-09] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Paired immunoglobulin-like type 2 receptors (PILRs) inhibitory PILRalpha and activating PILRbeta are predominantly expressed on myeloid cells. Their functions in host defense and inflammation are largely unknown, and in this study, we evaluated their roles in an acute Staphylococcus aureus pneumonia model. Compared to their respective controls, Pilrb(-/-) mice or mice in which PILRalpha was activated with an agonistic antibody showed improved clearance of pulmonary staphylococci and improved survival. These mice had reduced serum or bronchoalveolar lavage fluid levels of interleukin-1beta (IL-1beta), tumor necrosis factor alpha (TNF-alpha), and IL-6 and elevated levels of gamma interferon (IFN-gamma), IL-12, and IL-10. In contrast, mice in which PILRbeta was activated had increased lung bacterial burdens and higher mortality coupled with an intense proinflammatory response with highly elevated levels of IL-1beta, TNF-alpha, and IL-6. Treatment groups with reduced bacterial burdens had higher levels of Keratinocyte-derived chemokine (KC), macrophage inflammatory protein 2 (MIP-2), and MIP-1alpha in bronchoalveolar lavage fluid and an increased influx of neutrophils and macrophages to the lungs. Consistent with our in vivo findings, bone marrow-derived macrophages from Pilrb(-/-) mice released significantly less IL-1beta and TNF-alpha and more IFN-gamma and IL-12 than did the wild-type macrophages when directly stimulated with heat-killed S. aureus. To our knowledge, this is the first evidence that S. aureus directly interacts with PILRbeta. It provides a mechanism by which manipulating the balance in favor of an inhibitory PILR signal, by activation of PILRalpha or deletion of PILRbeta, helps to control acute S. aureus-mediated pneumonia and attenuate the inflammatory response. These results highlight the importance of PILRs in innate immunity and the control of inflammation.
Collapse
|
48
|
Whittaker GC, Orr SJ, Quigley L, Hughes L, Francischetti IMB, Zhang W, McVicar DW. The linker for activation of B cells (LAB)/non-T cell activation linker (NTAL) regulates triggering receptor expressed on myeloid cells (TREM)-2 signaling and macrophage inflammatory responses independently of the linker for activation of T cells. J Biol Chem 2009; 285:2976-85. [PMID: 19948717 DOI: 10.1074/jbc.m109.038398] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Triggering receptor expressed on myeloid cells-2 (TREM-2) is rapidly emerging as a key regulator of the innate immune response via its regulation of macrophage inflammatory responses. Here we demonstrate that proximal TREM-2 signaling parallels other DAP12-based receptor systems in its use of Syk and Src-family kinases. However, we find that the linker for activation of T cells (LAT) is severely reduced as monocytes differentiate into macrophages and that TREM-2 exclusively uses the linker for activation of B cells (LAB encoded by the gene Lat2(-/-)) to mediate downstream signaling. LAB is required for TREM-2-mediated activation of Erk1/2 and dampens proximal TREM-2 signals through a novel LAT-independent mechanism resulting in macrophages with proinflammatory properties. Thus, Lat2(-/-) macrophages have increased TREM-2-induced proximal phosphorylation, and lipopolysaccharide stimulation of these cells leads to increased interleukin-10 (IL-10) and decreased IL-12p40 production relative to wild type cells. Together these data identify LAB as a critical, LAT-independent regulator of TREM-2 signaling and macrophage development capable of controlling subsequent inflammatory responses.
Collapse
Affiliation(s)
- Gillian C Whittaker
- Cancer and Inflammation Program, Center for Cancer Research, NCI-Frederick, National Institutes of Health, Frederick, Maryland 21702, USA
| | | | | | | | | | | | | |
Collapse
|
49
|
Zhang X, Majlessi L, Deriaud E, Leclerc C, Lo-Man R. Coactivation of Syk kinase and MyD88 adaptor protein pathways by bacteria promotes regulatory properties of neutrophils. Immunity 2009; 31:761-71. [PMID: 19913447 DOI: 10.1016/j.immuni.2009.09.016] [Citation(s) in RCA: 242] [Impact Index Per Article: 15.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2009] [Revised: 06/20/2009] [Accepted: 09/09/2009] [Indexed: 12/31/2022]
Abstract
Neutrophils are one of the first lines of defense against microbial pathogens and are rapidly recruited at the infection site upon inflammatory conditions. We show here that after bacterial stimulation, and in contrast to monocytes and macrophages, murine neutrophils contributed poorly to inflammatory responses; however, they secreted high amounts of the anti-inflammatory cytokine IL-10 in a DAP12 adaptor-Syk kinase and MyD88 adaptor-dependent manner. Cotriggering of TLR-MyD88- and C-type lectin receptor (CLR)-Syk-dependent pathways led to a quick and sustained phosphorylation of p38 MAP and Akt kinases in neutrophils. In vivo, both Gram-negative bacteria and mycobacteria induced the recruitment of neutrophils secreting IL-10. In acute mycobacterial infection, neutrophil-derived IL-10 controlled the inflammatory response of dendritic cells, monocytes and macrophages in the lung. During a chronic infection, neutrophil depletion promoted inflammation and decreased the mycobacterial burden. Therefore, neutrophils can have a previously unsuspected regulatory role during acute and chronic microbial infections.
Collapse
Affiliation(s)
- Xiaoming Zhang
- Institut Pasteur, Unité Régulation Immunitaire et Vaccinologie, 25 rue du Dr. Roux, 75724 Paris Cedex 15, France
| | | | | | | | | |
Collapse
|
50
|
Blank U, Launay P, Benhamou M, Monteiro RC. Inhibitory ITAMs as novel regulators of immunity. Immunol Rev 2009; 232:59-71. [DOI: 10.1111/j.1600-065x.2009.00832.x] [Citation(s) in RCA: 132] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
|