1
|
Lin YY, Chang WH, Hsieh SL, Cheng IHJ. The deficient CLEC5A ameliorates the behavioral and pathological deficits via the microglial Aβ clearance in Alzheimer's disease mouse model. J Neuroinflammation 2024; 21:273. [PMID: 39443966 PMCID: PMC11515658 DOI: 10.1186/s12974-024-03253-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Accepted: 10/04/2024] [Indexed: 10/25/2024] Open
Abstract
BACKGROUND Alzheimer's disease (AD) is a neurodegenerative disease that causes cognitive dysfunction in older adults. One of the AD pathological factors, β-Amyloid (Aβ), triggers inflammatory responses and phagocytosis of microglia. C-type lectin domain family 5 member A (CLEC5A) induces over-reactive inflammatory responses in several virus infections. Yet, the role of CLEC5A in AD progression remains unknown. This study aimed to elucidate the contribution of CLEC5A to Aβ-induced microglial activation and behavioral deficits. METHODS The AD mouse model was crossed with Clec5a knockout mice for subsequent behavioral and pathological tests. The memory deficit was revealed by the Morris water maze, while the nociception abnormalities were examined by the von Frey filament and hotplate test. The Aβ deposition and microglia recruitment were identified by ELISA and immunohistochemistry. The inflammatory signals were identified by ELISA and western blotting. In the Clec5a knockdown microglial cell model and Clec5a knockout primary microglia, the microglial phagocytosis was revealed using the fluorescent-labeled Aβ. RESULTS The AD mice with Clec5a knockout improved Aβ-induced memory deficit and abnormal nociception. These mice have reduced Aβ deposition and increased microglia coverage surrounding the amyloid plaque, suggesting the involvement of CLEC5A in AD progression and Aβ clearance. Moreover, the phagocytosis was also increased in the Aβ-stressed Clec5a knockdown microglial cell lines and Clec5a knockout primary microglia. CONCLUSION The Clec5a knockout ameliorates AD-like deficits by modulating microglial Aβ clearance. This study implies that targeting microglial Clec5a could offer a promising approach to mitigate AD progression.
Collapse
MESH Headings
- Animals
- Lectins, C-Type/metabolism
- Lectins, C-Type/deficiency
- Lectins, C-Type/genetics
- Microglia/metabolism
- Microglia/pathology
- Alzheimer Disease/metabolism
- Alzheimer Disease/pathology
- Alzheimer Disease/genetics
- Mice
- Amyloid beta-Peptides/metabolism
- Disease Models, Animal
- Mice, Knockout
- Mice, Inbred C57BL
- Male
- Mice, Transgenic
- Maze Learning/physiology
- Phagocytosis
- Receptors, Cell Surface/metabolism
- Receptors, Cell Surface/deficiency
- Receptors, Cell Surface/genetics
Collapse
Affiliation(s)
- Yu-Yi Lin
- Institute of Brain Science, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Wen-Han Chang
- Institute of Brain Science, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Shie-Liang Hsieh
- Immunology Research Center, National Health Research Institutes, Zhunan, Taiwan.
- Institute of Clinical Medicine, Institute of Microbiology and Immunology, National Yang Ming Chiao Tung University, Taipei, Taiwan.
- Department of Medical Research, Taipei Veterans General Hospital, Taipei, Taiwan.
| | - Irene Han-Juo Cheng
- Institute of Brain Science, National Yang Ming Chiao Tung University, Taipei, Taiwan.
- Brain Research Center, National Yang Ming Chiao Tung University, Taipei, Taiwan.
| |
Collapse
|
2
|
Furuya H, Nguyen CT, Chan T, Marusina AI, Merleev AA, Garcia-Hernandez MDLL, Hsieh SL, Tsokos GC, Ritchlin CT, Tagkopoulos I, Maverakis E, Adamopoulos IE. IL-23 induces CLEC5A + IL-17A + neutrophils and elicit skin inflammation associated with psoriatic arthritis. J Autoimmun 2024; 143:103167. [PMID: 38301504 PMCID: PMC10981569 DOI: 10.1016/j.jaut.2024.103167] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Revised: 01/04/2024] [Accepted: 01/09/2024] [Indexed: 02/03/2024]
Abstract
IL-23-activation of IL-17 producing T cells is involved in many rheumatic diseases. Herein, we investigate the role of IL-23 in the activation of myeloid cell subsets that contribute to skin inflammation in mice and man. IL-23 gene transfer in WT, IL-23RGFP reporter mice and subsequent analysis with spectral cytometry show that IL-23 regulates early innate immune events by inducing the expansion of a myeloid MDL1+CD11b+Ly6G+ population that dictates epidermal hyperplasia, acanthosis, and parakeratosis; hallmark pathologic features of psoriasis. Genetic ablation of MDL-1, a major PU.1 transcriptional target during myeloid differentiation exclusively expressed in myeloid cells, completely prevents IL-23-pathology. Moreover, we show that IL-23-induced myeloid subsets are also capable of producing IL-17A and IL-23R+MDL1+ cells are present in the involved skin of psoriasis patients and gene expression correlations between IL-23 and MDL-1 have been validated in multiple patient cohorts. Collectively, our data demonstrate a novel role of IL-23 in MDL-1-myelopoiesis that is responsible for skin inflammation and related pathologies. Our data open a new avenue of investigations regarding the role of IL-23 in the activation of myeloid immunoreceptors and their role in autoimmunity.
Collapse
Affiliation(s)
- Hiroki Furuya
- Department of Rheumatology and Clinical Immunology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, USA
| | - Cuong Thach Nguyen
- Division of Rheumatology, Allergy and Clinical Immunology, University of California, Davis, USA
| | - Trevor Chan
- Department of Computer Science, University of California, Davis, CA, USA; Genome Center, University of California, Davis, CA, USA
| | - Alina I Marusina
- Department of Dermatology, University of California, Davis, Sacramento, USA
| | | | | | - Shie-Liang Hsieh
- Genomics Research Center, Academia Sinica, Nankang, Taipei, Taiwan
| | - George C Tsokos
- Division of Rheumatology, Allergy and Clinical Immunology, University of California, Davis, USA
| | - Christopher T Ritchlin
- Division of Allergy, Immunology & Rheumatology, University of Rochester Medical School, NY, USA
| | - Ilias Tagkopoulos
- Department of Computer Science, University of California, Davis, CA, USA; Process Integration and Predictive Analytics, PIPA LLC, CA, USA
| | - Emanual Maverakis
- Department of Dermatology, University of California, Davis, Sacramento, USA
| | - Iannis E Adamopoulos
- Department of Rheumatology and Clinical Immunology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, USA; Division of Rheumatology, Allergy and Clinical Immunology, University of California, Davis, USA.
| |
Collapse
|
3
|
Chen H, Chen E, Lu Y, Xu Y. Identification of immune-related genes in diagnosing retinopathy of prematurity with sepsis through bioinformatics analysis and machine learning. Front Genet 2023; 14:1264873. [PMID: 38028617 PMCID: PMC10667920 DOI: 10.3389/fgene.2023.1264873] [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: 08/15/2023] [Accepted: 10/30/2023] [Indexed: 12/01/2023] Open
Abstract
Background: There is increasing evidence indicating that immune system dysregulation plays a pivotal role in the pathogenesis of retinopathy of prematurity (ROP) and sepsis. This study aims to identify key diagnostic candidate genes in ROP with sepsis. Methods: We obtained publicly available data on ROP and sepsis from the gene expression omnibus database. Differential analysis and weighted gene correlation network analysis (WGCNA) were performed to identify differentially expressed genes (DEGs) and key module genes. Subsequently, we conducted functional enrichment analysis to gain insights into the biological functions and pathways. To identify immune-related pathogenic genes and potential mechanisms, we employed several machine learning algorithms, including Support Vector Machine Recursive Feature Elimination (SVM-RFE), Least Absolute Shrinkage and Selection Operator (LASSO), and Random Forest (RF). We evaluated the diagnostic performance using nomogram and Receiver Operating Characteristic (ROC) curves. Furthermore, we used CIBERSORT to investigate immune cell dysregulation in sepsis and performed cMAP analysis to identify potential therapeutic drugs. Results: The sepsis dataset comprised 352 DEGs, while the ROP dataset had 307 DEGs and 420 module genes. The intersection between DEGs for sepsis and module genes for ROP consisted of 34 genes, primarily enriched in immune-related pathways. After conducting PPI network analysis and employing machine learning algorithms, we pinpointed five candidate hub genes. Subsequent evaluation using nomograms and ROC curves underscored their robust diagnostic potential. Immune cell infiltration analysis revealed immune cell dysregulation. Finally, through cMAP analysis, we identified some small molecule compounds that have the potential for sepsis treatment. Conclusion: Five immune-associated candidate hub genes (CLEC5A, KLRB1, LCN2, MCEMP1, and MMP9) were recognized, and the nomogram for the diagnosis of ROP with sepsis was developed.
Collapse
Affiliation(s)
- Han Chen
- Department of Ophthalmology, School of Medicine, Xinhua Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Enguang Chen
- Department of Ophthalmology, School of Medicine, Xinhua Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Yao Lu
- Department of Ophthalmology, Anhui No. 2 Provincial People’s Hospital, Anhui, Hefei, China
| | - Yu Xu
- Department of Ophthalmology, School of Medicine, Xinhua Hospital, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
4
|
Suchanti S, Stephen BJ, Chaurasia TP, Raghuwanshi AP, Singh G, Singh A, Mishra R. In-Silico CLEC5A mRNA expression analysis to predict Dengue susceptibility in cancer patients. Biochem Biophys Rep 2023; 35:101501. [PMID: 37415850 PMCID: PMC10320400 DOI: 10.1016/j.bbrep.2023.101501] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2023] [Revised: 05/31/2023] [Accepted: 06/08/2023] [Indexed: 07/08/2023] Open
Abstract
Dengue fever is the fastest-growing infectious disease in the world. It is the leading vector-borne viral neglected tropical disease. The most acute immune response to dengue virus infection is dengue shock syndrome and hemorrhagic fever, which is due to the activation of CLEC5A C-type lectin domain family 5, member A (CLEC5A). It is a cell surface receptor, and its well-known ligand is the dengue virus. It gets activated by the attachment of dengue virion, which, as a result, phosphorylates its adaptor protein DAP12 leading to the induction of various pro-inflammatory cytokines. Clinical data suggested that the kidneys and lungs are among the major hit organs in the case of severe dengue infection. Here we predict kidney and lung cancer patients are vulnerable to dengue virus infection as CLEC5A mRNA expression in tumor samples using publicly available software such as TIMER and GEPIA database. We also identified the immunomodulatory role CLEC5A gene therefore targeting it could be a vital tool to cure dengue.
Collapse
Affiliation(s)
- Surabhi Suchanti
- Disease Biology Lab, Department of Life Sciences and Biotechnology, Chhatrapati Shahu Ji Maharaj University, Kanpur, UP, 208024, India
- Department of Biosciences, Manipal University Jaipur, Dehmi Kalan, Jaipur, 303007, Rajasthan, India
| | - Bjorn John Stephen
- Department of Biosciences, Manipal University Jaipur, Dehmi Kalan, Jaipur, 303007, Rajasthan, India
| | - Tejulal Prasad Chaurasia
- Department of Biosciences, Manipal University Jaipur, Dehmi Kalan, Jaipur, 303007, Rajasthan, India
- School of Allied Health Sciences, Jaipur National University, Jaipur, Rajasthan, 302017, India
| | - Amit Prakash Raghuwanshi
- Department of Botany, Dayanand Anglo - Vedic (PG) College, Civil Lines, Kanpur, 208001, UP, India
| | - Gyanendra Singh
- Toxicology Department, ICMR-National Institute of Occupational Health, Ahmedabad, 380016, India
| | - Abhijeet Singh
- Department of Biosciences, Manipal University Jaipur, Dehmi Kalan, Jaipur, 303007, Rajasthan, India
| | - Rajeev Mishra
- Disease Biology Lab, Department of Life Sciences and Biotechnology, Chhatrapati Shahu Ji Maharaj University, Kanpur, UP, 208024, India
| |
Collapse
|
5
|
Molecular Mechanism and Role of Japanese Encephalitis Virus Infection in Central Nervous System-Mediated Diseases. Viruses 2022; 14:v14122686. [PMID: 36560690 PMCID: PMC9781168 DOI: 10.3390/v14122686] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Revised: 11/21/2022] [Accepted: 11/25/2022] [Indexed: 12/05/2022] Open
Abstract
The Japanese encephalitis virus (JEV) is the most common cause of neurodegenerative disease in Southeast Asia and the Western Pacific region; approximately 1.15 billion people are at risk, and thousands suffer from permanent neurological disorders across Asian countries, with 10-15 thousand people dying each year. JEV crosses the blood-brain barrier (BBB) and forms a complex with receptors on the surface of neurons. GRP78, Src, TLR7, caveolin-1, and dopamine receptor D2 are involved in JEV binding and entry into the neurons, and these receptors also play a role in carcinogenic activity in cells. JEV binds to GRP78, a member of the HSP70 overexpressed on malignant cells to enter neurons, indicating a higher chance of JEV infection in cancer patients. However, JEV enters human brain microvascular endothelial cells via an endocytic pathway mediated by caveolae and the ezrin protein and also targets dopamine-rich areas for infection of the midbrain via altering dopamine levels. In addition, JEV complexed with CLEC5A receptor of macrophage cells is involved in the breakdown of the BBB and central nervous system (CNS) inflammation. CLEC5A-mediated infection is also responsible for the influx of cytokines into the CNS. In this review, we discuss the neuronal and macrophage surface receptors involved in neuronal death.
Collapse
|
6
|
Chen R, Wu W, Chen SY, Liu ZZ, Wen ZP, Yu J, Zhang LB, Liu Z, Zhang J, Luo P, Zeng WJ, Cheng Q. A Pan-Cancer Analysis Reveals CLEC5A as a Biomarker for Cancer Immunity and Prognosis. Front Immunol 2022; 13:831542. [PMID: 35979347 PMCID: PMC9376251 DOI: 10.3389/fimmu.2022.831542] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Accepted: 05/05/2022] [Indexed: 11/17/2022] Open
Abstract
Background CLEC5A is a member of the C-type lectin superfamily. It can activate macrophages and lead to a series of immune-inflammation reactions. Previous studies reveal the role of CLEC5A in infection and inflammation diseases. Method We acquire and analyze data from The Cancer Genome Atlas (TCGA) database, Genotype-Tissue Expression (GTEx) database, and other comprehensive databases via GSCALite, cBioPortal, and TIMER 2.0 platforms or software. Single-cell sequencing analysis was performed for quantifying the tumor microenvironment of several types of cancers. Results CLEC5A is differentially expressed in a few cancer types, of which overexpression accompanies low overall survival of patients. DNA methylation mainly negatively correlates with CLEC5A expression. Moreover, CLEC5A is positively related to immune infiltration, including macrophages, cancer-associated fibroblasts (CAFs), and regulatory T cells (Tregs). Immune checkpoint genes are significantly associated with CLEC5A expression in diverse cancers. In addition, CLEC5A expression correlates with mismatch repair (MMR) in several cancers. Tumor mutation burden (TMB), microsatellite instability (MSI), and neoantigens show a positive association with CLEC5A expression in several cancers. Furthermore, CLEC5A in cancer correlates with signal transduction, the immune system, EMT, and apoptosis process. The drug sensitivity analysis screens out potential therapeutic agents associated with CLEC5A expression, including FR-180204, Tivozanib, OSI-930, Linifanib, AC220, VNLG/124, Bexarotene, omacetaxine mepesuccinate, narciclasine, leptomycin B, PHA-793887, LRRK2-IN-1, and CR-1-31B. Conclusion CLEC5A overexpresses in multiple cancers in contrast to normal tissues, and high CLEC5A expression predicts poor prognosis of patients and immune infiltration. CLEC5A is a potential prognostic biomarker of diverse cancers and a target for anti-tumor therapy.
Collapse
Affiliation(s)
- Rui Chen
- Department of Neurosurgery, The Affiliated Nanhua Hospital, Hengyang Medical School, University of South China, Hengyang, China
| | - Wantao Wu
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Si-Yu Chen
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha, China
| | - Zheng-Zheng Liu
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Zhi-Peng Wen
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha, China
| | - Jing Yu
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
- Department of Pharmacy, The Affiliated Hospital of Guizhou Medical University, Guizhou Medical University, Guiyang, China
| | - Long-Bo Zhang
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
- Department of Neurosurgery, and Department of Cellular & Molecular Physiology, Yale University School of Medicine, New Haven, CT, United States
| | - Zaoqu Liu
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Jian Zhang
- Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Peng Luo
- Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Wen-Jing Zeng
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha, China
- *Correspondence: Quan Cheng, ; Wen-Jing Zeng,
| | - Quan Cheng
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
- *Correspondence: Quan Cheng, ; Wen-Jing Zeng,
| |
Collapse
|
7
|
Peek CT, Ford CA, Eichelberger KR, Jacobse J, Torres TP, Maseda D, Latour YL, Piazuelo MB, Johnson JR, Byndloss MX, Wilson KT, Rathmell JC, Goettel JA, Cassat JE. Intestinal Inflammation Promotes MDL-1 + Osteoclast Precursor Expansion to Trigger Osteoclastogenesis and Bone Loss. Cell Mol Gastroenterol Hepatol 2022; 14:731-750. [PMID: 35835390 PMCID: PMC9420375 DOI: 10.1016/j.jcmgh.2022.07.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/26/2022] [Revised: 07/01/2022] [Accepted: 07/05/2022] [Indexed: 12/13/2022]
Abstract
BACKGROUND & AIMS Inflammatory bowel disease (IBD) is characterized by severe gastrointestinal inflammation, but many patients experience extra-intestinal disease. Bone loss is one common extra-intestinal manifestation of IBD that occurs through dysregulated interactions between osteoclasts and osteoblasts. Systemic inflammation has been postulated to contribute to bone loss, but the specific pathologic mechanisms have not yet been fully elucidated. We hypothesized that intestinal inflammation leads to bone loss through increased abundance and altered function of osteoclast progenitors. METHODS We used chemical, T cell driven, and infectious models of intestinal inflammation to determine the impact of intestinal inflammation on bone volume, the skeletal cytokine environment, and the cellular changes to pre-osteoclast populations within bone marrow. Additionally, we evaluated the potential for monoclonal antibody treatment against an inflammation-induced osteoclast co-receptor, myeloid DNAX activation protein 12-associating lectin-1 (MDL-1) to reduce bone loss during colitis. RESULTS We observed significant bone loss across all models of intestinal inflammation. Bone loss was associated with an increase in pro-osteoclastogenic cytokines within the bone and an expansion of a specific Cd11b-/loLy6Chi osteoclast precursor (OCP) population. Intestinal inflammation led to altered OCP expression of surface receptors involved in osteoclast differentiation and function, including the pro-osteoclastogenic co-receptor MDL-1. OCPs isolated from mice with intestinal inflammation demonstrated enhanced osteoclast differentiation ex vivo compared to controls, which was abrogated by anti-MDL-1 antibody treatment. Importantly, in vivo anti-MDL-1 antibody treatment ameliorated bone loss during intestinal inflammation. CONCLUSIONS Collectively, these data implicate the pathologic expansion and altered function of OCPs expressing MDL-1 in bone loss during IBD.
Collapse
Affiliation(s)
- Christopher T Peek
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Caleb A Ford
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee
| | - Kara R Eichelberger
- Department of Pediatrics, Division of Pediatric Infectious Diseases, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Justin Jacobse
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee; Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Teresa P Torres
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Damian Maseda
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee; Department of Dermatology, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Yvonne L Latour
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee
| | - M Blanca Piazuelo
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee; Center for Mucosal Inflammation and Cancer, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Joshua R Johnson
- Vanderbilt Center for Bone Biology, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Mariana X Byndloss
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee; Vanderbilt Institute for Infection, Immunology, and Inflammation, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Keith T Wilson
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee; Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee; Vanderbilt Institute for Infection, Immunology, and Inflammation, Vanderbilt University Medical Center, Nashville, Tennessee; Center for Mucosal Inflammation and Cancer, Vanderbilt University Medical Center, Nashville, Tennessee; Veterans Affairs Tennessee Valley Healthcare System, Nashville, Tennessee
| | - Jeffrey C Rathmell
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee; Vanderbilt Institute for Infection, Immunology, and Inflammation, Vanderbilt University Medical Center, Nashville, Tennessee; Center for Mucosal Inflammation and Cancer, Vanderbilt University Medical Center, Nashville, Tennessee; Vanderbilt Center for Immunobiology, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Jeremy A Goettel
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee; Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee; Vanderbilt Institute for Infection, Immunology, and Inflammation, Vanderbilt University Medical Center, Nashville, Tennessee; Center for Mucosal Inflammation and Cancer, Vanderbilt University Medical Center, Nashville, Tennessee
| | - James E Cassat
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee; Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee; Department of Pediatrics, Division of Pediatric Infectious Diseases, Vanderbilt University Medical Center, Nashville, Tennessee; Vanderbilt Institute for Infection, Immunology, and Inflammation, Vanderbilt University Medical Center, Nashville, Tennessee; Center for Mucosal Inflammation and Cancer, Vanderbilt University Medical Center, Nashville, Tennessee; Vanderbilt Center for Immunobiology, Vanderbilt University Medical Center, Nashville, Tennessee; Vanderbilt Center for Bone Biology, Vanderbilt University Medical Center, Nashville, Tennessee.
| |
Collapse
|
8
|
Chen PK, Wey SJ, Chen DY. Interleukin-18: a biomarker with therapeutic potential in adult-onset Still's disease. Expert Rev Clin Immunol 2022; 18:823-833. [PMID: 35771972 DOI: 10.1080/1744666x.2022.2096592] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
INTRODUCTION Adult-onset Still's disease (AOSD) is an autoinflammatory disease driven by the innate immune response. Given the ambiguity in clinical presentation and lack of specific diagnostic biomarkers, AOSD diagnosis is usually delayed in the early stage. Because AOSD is a rare disease with clinical heterogeneity, there is no consensus on its treatment currently. This review summarizes the current research evidence regarding the pathogenic role and the diagnostic or therapeutic potential of interleukin (IL)-18 in AOSD. AREAS COVERED We searched the MEDLINE database using the PubMed interface and reviewed English-language literature from 1971 to 2022. This review focusing on IL-18 discusses its pathogenic role and clinical implications in AOSD. EXPERT OPINION NLRP3-inflammasome activation with IL-18 overproduction plays a pathogenic role in AOSD. IL-18 is closely linked to the clinical manifestations and disease activity of AOSD and may be a diagnostic biomarker. Given its pathogenic role in AOSD, IL-18 could become a potential therapeutic target. IL-18 binding protein (IL-18BP) negatively regulates the biological activity of IL-18 by inhibiting IL-18 signaling, and a clinical trial revealed that IL-18BP (Tadekinig alfa) treatment was well-tolerated and effective for AOSD. Recently, monoclonal antibodies against IL-18 have been under evaluation in a phase 1b trial.
Collapse
Affiliation(s)
- Po-Ku Chen
- Rheumatology and Immunology Center, China Medical University Hospital, Taichung, Taiwan.,College of Medicine, China Medical University, Taichung, Taiwan.,Translational Medicine Laboratory, Rheumatology and Immunology Center, Taichung, Taiwan
| | - Shiow-Jiuan Wey
- Division of Dermatology, Chung Shan Medical University Hospital, Taichung, Taiwan.,Institute of Medicine, Chung Shan Medical University Hospital, Taichung, Taiwan
| | - Der-Yuan Chen
- Rheumatology and Immunology Center, China Medical University Hospital, Taichung, Taiwan.,College of Medicine, China Medical University, Taichung, Taiwan.,Translational Medicine Laboratory, Rheumatology and Immunology Center, Taichung, Taiwan.,Institute of Medicine, Chung Shan Medical University Hospital, Taichung, Taiwan.,D. Program in Translational Medicine and Rong Hsing Research Center for Translational Medicine, National Chung Hsing UniversityPh., Taichung, Taiwan
| |
Collapse
|
9
|
Activation of the Innate Immune Checkpoint CLEC5A on Myeloid Cells in the Absence of Danger Signals Modulates Macrophages’ Function but Does Not Trigger the Adaptive T Cell Immune Response. J Immunol Res 2022; 2022:9926305. [PMID: 35252461 PMCID: PMC8896916 DOI: 10.1155/2022/9926305] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Revised: 11/30/2021] [Accepted: 02/09/2022] [Indexed: 12/19/2022] Open
Abstract
C-Type lectin receptor 5A (CLEC5A) is a spleen tyrosine kinase- (Syk-) coupled pattern recognition receptor expressed on myeloid cells and involved in the innate immune response to viral and bacterial infections. Activation of the CLEC5A receptor with pathogen-derived antigens leads to a secretion of proinflammatory mediators such as TNF-α and IL-6 that may provoke a systemic cytokine storm, and CLEC5A gene polymorphisms are associated with the severity of DV infection. In addition, the CLEC5A receptor was mentioned in the context of noninfectious disorders like chronic obstructive pulmonary disease (COPD) or arthritis. Altogether, CLEC5A may be considered as an innate immune checkpoint capable to amplify proinflammatory signals, and this way contributes to infection or to aseptic inflammation. In this study, we determined CLEC5A receptor expression on different macrophage subsets (in vitro and ex vivo) and the functional consequences of its activation in aseptic conditions. The CLEC5A surface expression appeared the highest on proinflammatory M1 macrophages while intermediate on tumor-associated phenotypes (M2c or TAM). In contrast, the CLEC5A expression on ex vivo-derived alveolar macrophages from healthy donors or macrophages from ovarian cancer patients was hardly detectable. Targeting CLEC5A on noninflammatory macrophages with an agonistic α-CLEC5A antibody triggered a release of proinflammatory cytokines, resembling a response to dengue virus, and led to phenotypic changes in myeloid cells that may suggest their reprogramming towards a proinflammatory phenotype, e.g., upregulation of CD80 and downregulation of CD163. Interestingly, the CLEC5A agonist upregulated immune-regulatory molecules like CD206, PD-L1, and cytokines like IL-10, macrophage-derived chemokine (MDC/CCL22), and thymus and activation chemokine (TARC/CCL17) which are associated with an anti-inflammatory or a protumorigenic macrophage phenotype. In the absence of concomitant pathogenic or endogenous danger signals, the CLEC5A receptor activation did not amplify an autologous T cell response, which may represent a protective innate mechanism to avoid an undesirable autoimmune adaptive response.
Collapse
|
10
|
Rossi ÁD, Higa LM, Herlinger AL, Ribeiro-Alves M, de Menezes MT, Giannini ALM, Cardoso CC, Da Poian AT, Tanuri A, Aguiar RS. Differential Expression of Human MicroRNAs During Dengue Virus Infection in THP-1 Monocytes. Front Cell Infect Microbiol 2021; 11:714088. [PMID: 34568093 PMCID: PMC8455953 DOI: 10.3389/fcimb.2021.714088] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Accepted: 08/25/2021] [Indexed: 12/31/2022] Open
Abstract
Dengue virus (DENV) is the most widespread arbovirus, responsible for a wide range of clinical manifestations, varying from self-limited illness to severe hemorrhagic fever. Dengue severity is associated with host intense proinflammatory response and monocytes have been considered one of the key cell types involved in the early steps of DENV infection and immunopathogenesis. To better understand cellular mechanisms involved in monocyte infection by DENV, we analyzed the expression levels of 754 human microRNAs in DENV-infected THP-1 cells, a human monocytic cell line. Eleven human microRNAs showed differential expression after DENV infection and gene ontology and enrichment analysis revealed biological processes potentially affected by these molecules. Five downregulated microRNAs were significantly linked to cellular response to stress, four to cell death/apoptosis, two to innate immune responses and one upregulated to vesicle mediated, TGF-β signaling, phosphatidylinositol mediated signaling, lipid metabolism process and blood coagulation.
Collapse
Affiliation(s)
- Átila Duque Rossi
- Laboratório de Virologia Molecular, Departamento de Genética, Instituto de Biologia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Luiza Mendonça Higa
- Laboratório de Virologia Molecular, Departamento de Genética, Instituto de Biologia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil.,Laboratório de Bioquímica de Vírus, Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Alice Laschuk Herlinger
- Laboratório de Virologia Molecular, Departamento de Genética, Instituto de Biologia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Marcelo Ribeiro-Alves
- Laboratório de Pesquisa Clínica em DST/AIDS, Instituto Nacional de Infectologia Evandro Chagas, FIOCRUZ, Rio de Janeiro, Brazil
| | - Mariane Talon de Menezes
- Laboratório de Virologia Molecular, Departamento de Genética, Instituto de Biologia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Ana Lucia Moraes Giannini
- Laboratório de Genômica Funcional e Transdução de Sinal, Departamento de Genética, Instituto de Biologia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Cynthia Chester Cardoso
- Laboratório de Virologia Molecular, Departamento de Genética, Instituto de Biologia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Andrea T Da Poian
- Laboratório de Bioquímica de Vírus, Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Amilcar Tanuri
- Laboratório de Virologia Molecular, Departamento de Genética, Instituto de Biologia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Renato Santana Aguiar
- Laboratório de Virologia Molecular, Departamento de Genética, Instituto de Biologia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil.,Laboratório de Biologia Integrativa, Departamento de Genética, Ecologia e Evolução, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| |
Collapse
|
11
|
Sung PS, Hsieh SL. C-type lectins and extracellular vesicles in virus-induced NETosis. J Biomed Sci 2021; 28:46. [PMID: 34116654 PMCID: PMC8193014 DOI: 10.1186/s12929-021-00741-7] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Accepted: 06/08/2021] [Indexed: 12/12/2022] Open
Abstract
Dysregulated formation of neutrophil extracellular traps (NETs) is observed in acute viral infections. Moreover, NETs contribute to the pathogenesis of acute viral infections, including those caused by the dengue virus (DV) and severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2). Furthermore, excessive NET formation (NETosis) is associated with disease severity in patients suffering from SARS-CoV-2-induced multiple organ injuries. Dendritic cell-specific intercellular adhesion molecule-3-grabbing non-integrin (DC-SIGN) and other members of C-type lectin family (L-SIGN, LSECtin, CLEC10A) have been reported to interact with viral glycans to facilitate virus spreading and exacerbates inflammatory reactions. Moreover, spleen tyrosine kinase (Syk)-coupled C-type lectin member 5A (CLEC5A) has been shown as the pattern recognition receptor for members of flaviviruses, and is responsible for DV-induced cytokine storm and Japanese encephalomyelitis virus (JEV)-induced neuronal inflammation. Moreover, DV activates platelets via CLEC2 to release extracellular vesicles (EVs), including microvesicles (MVs) and exosomes (EXOs). The DV-activated EXOs (DV-EXOs) and MVs (DV-MVs) stimulate CLEC5A and Toll-like receptor 2 (TLR2), respectively, to enhance NET formation and inflammatory reactions. Thus, EVs from virus-activated platelets (PLT-EVs) are potent endogenous danger signals, and blockade of C-type lectins is a promising strategy to attenuate virus-induced NETosis and intravascular coagulopathy.
Collapse
Affiliation(s)
- Pei-Shan Sung
- Genomics Research Center, Academia Sinica, 128, Academia Road, Sec. 2, Nankang District, Taipei, 115 Taiwan
| | - Shie-Liang Hsieh
- Genomics Research Center, Academia Sinica, 128, Academia Road, Sec. 2, Nankang District, Taipei, 115 Taiwan
- Institute of Clinical Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Department of Medical Research, Taipei Veterans General Hospital, Taipei, Taiwan
- Institute for Cancer Biology and Drug Discovery, Taipei Medical University, Taipei, Taiwan
- Institute of Immunology, College of Medicine, National Taiwan University, Taipei, Taiwan
| |
Collapse
|
12
|
Menegatti S, Guillemot V, Latis E, Yahia-Cherbal H, Mittermüller D, Rouilly V, Mascia E, Rosine N, Koturan S, Millot GA, Leloup C, Duffy D, Gleizes A, Hacein-Bey-Abina S, Sellam J, Berenbaum F, Miceli-Richard C, Dougados M, Bianchi E, Rogge L. Immune response profiling of patients with spondyloarthritis reveals signalling networks mediating TNF-blocker function in vivo. Ann Rheum Dis 2021; 80:475-486. [PMID: 33268443 PMCID: PMC7958106 DOI: 10.1136/annrheumdis-2020-218304] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Revised: 10/30/2020] [Accepted: 10/30/2020] [Indexed: 12/24/2022]
Abstract
OBJECTIVES Antitumour necrosis factor (TNF) therapy has revolutionised treatment of several chronic inflammatory diseases, including spondyloarthritis (SpA). However, TNF inhibitors (TNFi) are not effective in all patients and the biological basis for treatment failure remains unknown. We have analysed induced immune responses to define the mechanism of action of TNF blockers in SpA and to identify immunological correlates of responsiveness to TNFi. METHODS Immune responses to microbial and pathway-specific stimuli were analysed in peripheral blood samples from 80 patients with axial SpA before and after TNFi treatment, using highly standardised whole-blood stimulation assays. Cytokines and chemokines were measured in a Clinical Laboratory Improvement Amendments (CLIA)-certified laboratory, and gene expression was monitored using nCounter assays. RESULTS Anti-TNF therapy induced profound changes in patients' innate immune responses. TNFi action was selective, and had only minor effects on Th1/Th17 immunity. Modular transcriptional repertoire analysis identified prostaglandin E2 synthesis and signalling, leucocyte recirculation, macrophage polarisation, dectin and interleukin (IL)-1 signalling, as well as the nuclear factor kappa B (NF-kB) transcription factor family as key pathways targeted by TNF blockers in vivo. Analysis of induced immune responses before treatment initiation revealed that expression of molecules associated with leucocyte adhesion and invasion, chemotaxis and IL-1 signalling are correlated with therapeutic responses to anti-TNF. CONCLUSIONS We show that TNFi target multiple immune cell pathways that cooperate to resolve inflammation. We propose that immune response profiling provides new insight into the biology of TNF-blocker action in patients and can identify signalling pathways associated with therapeutic responses to biological therapies.
Collapse
Affiliation(s)
- Silvia Menegatti
- Immunoregulation Unit, Department of Immunology, Institut Pasteur, Paris, France
- Université Paris Diderot, Sorbonne Paris Cité, Paris, France
- INSERM U932, Institut Curie, PSL Research University, Paris, France
| | - Vincent Guillemot
- Bioinformatics and Biostatistics Hub-Département de Biologie Computationelle, Institut Pasteur, USR 3756 IP CNRS, Paris, France
| | - Eleonora Latis
- Immunoregulation Unit, Department of Immunology, Institut Pasteur, Paris, France
- Université Paris Diderot, Sorbonne Paris Cité, Paris, France
| | - Hanane Yahia-Cherbal
- Immunoregulation Unit, Department of Immunology, Institut Pasteur, Paris, France
- Université Paris Diderot, Sorbonne Paris Cité, Paris, France
| | - Daniela Mittermüller
- Immunoregulation Unit, Department of Immunology, Institut Pasteur, Paris, France
| | | | - Elena Mascia
- Immunoregulation Unit, Department of Immunology, Institut Pasteur, Paris, France
| | - Nicolas Rosine
- Immunoregulation Unit, Department of Immunology, Institut Pasteur, Paris, France
- Université Paris Diderot, Sorbonne Paris Cité, Paris, France
| | - Surya Koturan
- Immunoregulation Unit, Department of Immunology, Institut Pasteur, Paris, France
- Université Paris Diderot, Sorbonne Paris Cité, Paris, France
| | - Gael A Millot
- Bioinformatics and Biostatistics Hub-Département de Biologie Computationelle, Institut Pasteur, USR 3756 IP CNRS, Paris, France
| | - Claire Leloup
- Immunoregulation Unit, Department of Immunology, Institut Pasteur, Paris, France
| | - Darragh Duffy
- Institut Pasteur, Translational Immunology Laboratory, Department of Immunology, Paris, France
| | - Aude Gleizes
- Clinical Immunology Laboratory, Groupe Hospitalier Universitaire Paris-Sud, Hôpital Kremlin-Bicêtre, AP-HP, Le-Kremlin-Bicêtre, France
- UTCBS CNRS UMR 8258, INSERM U1267, Faculté de Pharmacie de Paris, Université de Paris, Paris, France
| | - Salima Hacein-Bey-Abina
- Clinical Immunology Laboratory, Groupe Hospitalier Universitaire Paris-Sud, Hôpital Kremlin-Bicêtre, AP-HP, Le-Kremlin-Bicêtre, France
- UTCBS CNRS UMR 8258, INSERM U1267, Faculté de Pharmacie de Paris, Université de Paris, Paris, France
| | - Jérémie Sellam
- Sorbonne Université, Service de Rhumatologie, Hôpital Saint-Antoine, AP-HP, Paris, France
- Centre de Recherche Saint-Antoine, INSERM UMR_S 938, Paris, France
| | - Francis Berenbaum
- Sorbonne Université, Service de Rhumatologie, Hôpital Saint-Antoine, AP-HP, Paris, France
- Centre de Recherche Saint-Antoine, INSERM UMR_S 938, Paris, France
| | - Corinne Miceli-Richard
- Immunoregulation Unit, Department of Immunology, Institut Pasteur, Paris, France
- Paris Descartes University, Rheumatology Department, Cochin Hospital, AP-HP, Paris, France
- Unité Mixte AP-HP/Institut Pasteur, Institut Pasteur, Paris, France
| | - Maxime Dougados
- Paris Descartes University, Rheumatology Department, Cochin Hospital, AP-HP, Paris, France
- Unité Mixte AP-HP/Institut Pasteur, Institut Pasteur, Paris, France
- INSERM U1153 Clinical Epidemiology and Biostatistics, PRES Sorbonne Paris-Cité, Paris, France
| | - Elisabetta Bianchi
- Immunoregulation Unit, Department of Immunology, Institut Pasteur, Paris, France
- Unité Mixte AP-HP/Institut Pasteur, Institut Pasteur, Paris, France
| | - Lars Rogge
- Immunoregulation Unit, Department of Immunology, Institut Pasteur, Paris, France
- Unité Mixte AP-HP/Institut Pasteur, Institut Pasteur, Paris, France
| |
Collapse
|
13
|
Girón-Ulloa A, González-Domínguez E, Klimek RS, Patiño-Martínez E, Vargas-Ayala G, Segovia-Gamboa NC, Campos-Peña V, Rodríguez-Arellano ME, Meraz-Ríos MA, Campos-Campos SF, Sánchez-Torres C. Specific macrophage subsets accumulate in human subcutaneous and omental fat depots during obesity. Immunol Cell Biol 2020; 98:868-882. [PMID: 32696992 DOI: 10.1111/imcb.12380] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2019] [Revised: 07/18/2020] [Accepted: 07/19/2020] [Indexed: 12/17/2022]
Abstract
Obesity is a chronic inflammatory disease associated with adipose tissue macrophage (ATM) activation. ATMs from lean mice contribute to tissue homeostasis by their M2-oriented polarization, whereas obesity leads to an increase of M1 inflammatory ATMs that underlies obesity-related metabolic disorders. In humans, studies characterizing ATMs and their functional status are limited. Here we investigated ATM phenotype in visceral (VAT) and subcutaneous (SAT) adipose tissue from healthy lean and obese individuals using two molecules previously identified as markers of M1-like and M2-like/tissue-resident macrophages, the C-type lectin CLEC5A and the scavenger receptor CD163L1, respectively. CD163L1 was expressed by the majority of ATMs, and CD163L1+ ATM density was greater with respect to cells expressing the pan-macrophage markers CD68 or CD11b. ATM counts in SAT, but not in VAT, increased in obese compared to lean individuals, measured with the three markers. Accordingly, CD163L1, CD68 and ITGAM gene expression was significantly enhanced in obese with respect to control individuals only in SAT. CLEC5A+ ATMs had a proinflammatory profile and were abundant in the lean VAT, but their density diminished in obesity. The only ATM subset that increased its counts in the obese VAT had a mixed M1-like (CD11c+ CD163- CD209- ) and M2-like (CLEC5A- CD206+ ) phenotype. ATM expansion was dominated by a subset of M2-like macrophages (CD11c- CLEC5A- CD163+ CD206+ CD209+ ) in the obese SAT, with a minor contribution of a CD11c+ CLEC5A- ATM subpopulation. Thus, both SAT and VAT seems to limit inflammation during obesity by differentially altering their ATM subset composition.
Collapse
Affiliation(s)
- Angélica Girón-Ulloa
- Department of Molecular Biomedicine, Centro de Investigación y de Estudios Avanzados del I.P.N., Mexico City, Mexico
| | - Erika González-Domínguez
- Department of Molecular Biomedicine, Centro de Investigación y de Estudios Avanzados del I.P.N., Mexico City, Mexico
| | - Rebeca S Klimek
- Department of Molecular Biomedicine, Centro de Investigación y de Estudios Avanzados del I.P.N., Mexico City, Mexico
| | - Eduardo Patiño-Martínez
- Department of Molecular Biomedicine, Centro de Investigación y de Estudios Avanzados del I.P.N., Mexico City, Mexico
| | - Germán Vargas-Ayala
- Department of Internal Medicine, Hospital General Ticomán, Mexico City, Mexico
| | - Norma C Segovia-Gamboa
- Department of Molecular Biomedicine, Centro de Investigación y de Estudios Avanzados del I.P.N., Mexico City, Mexico
| | - Victoria Campos-Peña
- Experimental Laboratory for Neurodegenerative Diseases, Instituto Nacional de Neurología y Neurocirugía "Manuel Velazco Suárez", Mexico City, Mexico
| | | | - Marco A Meraz-Ríos
- Department of Molecular Biomedicine, Centro de Investigación y de Estudios Avanzados del I.P.N., Mexico City, Mexico
| | | | - Carmen Sánchez-Torres
- Department of Molecular Biomedicine, Centro de Investigación y de Estudios Avanzados del I.P.N., Mexico City, Mexico
| |
Collapse
|
14
|
Elevated Expression of C-Type Lectin Domain Family 5-Member A (CLEC5A) and Its Relation to Inflammatory Parameters and Disease Course in Adult-Onset Still's Disease. J Immunol Res 2020; 2020:9473497. [PMID: 32377540 PMCID: PMC7195645 DOI: 10.1155/2020/9473497] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Accepted: 04/07/2020] [Indexed: 12/15/2022] Open
Abstract
C-type lectin domain family 5-member A (CLEC5A) associates with adaptor DAP12 (DNAX activation protein 12) to form receptor complexes involved in inflammatory responses. We postulated a potential role of CLEC5A in the pathogenesis of adult-onset Still's disease (AOSD) and aimed to investigate CLEC5A expression and its association with activity parameters and disease course. In 34 AOSD patients and 12 healthy controls (HC), circulating levels of CLEC5A-expressing monocytes or granulocytes were determined by flow cytometry analysis, the mRNA expression of CLEC5A and DAP12 on PBMCs by quantitative PCR, and plasma levels of proinflammatory cytokines by ELISA. AOSD patients had significantly higher percentages and mean fluorescence intensity (MFI) of CLEC5A-expressing monocytes (median 62.1% and 3.20, respectively) or granulocytes (72.6% and 3.22, respectively) compared with HC (in monocytes: 17.0% and 0.65, both p < 0.001; in granulocytes: 67.3%, p < 0.05 and 0.90, p < 0.001; respectively). Patients also had significantly higher levels of CLEC5A mRNA expression on PBMCs compared with HC (median 1.77 vs. 0.68, p < 0.05). The levels of CLEC5A-expressing monocytes or granulocytes were positively associated with activity scores and levels of IL-1β and IL-18 in AOSD patients. The patients with a systemic pattern had significantly higher levels of CLEC5A-expressing granulocytes and IL-18 compared to those with a chronic articular pattern of disease course. After 6 months of therapy, levels of CLEC5A-expressing monocytes and granulocytes significantly declined, paralleling the decrease of AOSD activity. Elevated CLEC5A levels and their positive association with activity parameters suggest that CLEC5A is involved in the pathogenesis and may serve as an activity indicator of AOSD.
Collapse
|
15
|
Sung PS, Chang WC, Hsieh SL. CLEC5A: A Promiscuous Pattern Recognition Receptor to Microbes and Beyond. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1204:57-73. [PMID: 32152943 PMCID: PMC7121389 DOI: 10.1007/978-981-15-1580-4_3] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
CLEC5A is a spleen tyrosine kinase (Syk)-coupled C-type lectin that is highly expressed by monocytes, macrophages, neutrophils, and dendritic cells and interacts with virions directly, via terminal fucose and mannose moieties of viral glycans. CLEC5A also binds to N-acetylglucosamine (GlcNAc) and N-acetylmuramic acid (MurNAc) disaccharides of bacterial cell walls. Compared to other C-type lectins (DC-SIGN and DC-SIGNR) and TLRs, CLEC5A binds its ligands with relatively low affinities. However, CLEC5A forms a multivalent hetero-complex with DC-SIGN and other C-type lectins upon engagement with ligands, and thereby mediates microbe-induced inflammatory responses via activation of Syk. For example, in vivo studies in mouse models have demonstrated that CLEC5A is responsible for flaviviruses-induced hemorrhagic shock and neuroinflammation, and a CLEC5A polymorphism in humans is associated with disease severity following infection with dengue virus. In addition, CLEC5A is co-activated with TLR2 by Listeria and Staphylococcus. Furthermore, CLEC5A-postive myeloid cells are responsible for Concanavilin A-induced aseptic inflammatory reactions. Thus, CLEC5A is a promiscuous pattern recognition receptor in myeloid cells and is a potential therapeutic target for attenuation of both septic and aseptic inflammatory reactions.
Collapse
Affiliation(s)
- Pei-Shan Sung
- Genomics Research Center, Academia Sinica, Taipei, Taiwan
| | - Wei-Chiao Chang
- School of Pharmacy, Taipei Medical University, Taipei, Taiwan
| | - Shie-Liang Hsieh
- Genomics Research Center, Academia Sinica, Taipei, Taiwan. .,School of Medicine, Institute of Clinical Medicine, National Yang-Ming University, Taipei, Taiwan.
| |
Collapse
|
16
|
Yoshikawa FSY, Pietrobon AJ, Branco ACCC, Pereira NZ, Oliveira LMDS, Machado CM, Duarte AJDS, Sato MN. Zika Virus Infects Newborn Monocytes Without Triggering a Substantial Cytokine Response. J Infect Dis 2020; 220:32-40. [PMID: 30785182 DOI: 10.1093/infdis/jiz075] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2019] [Accepted: 02/13/2019] [Indexed: 12/18/2022] Open
Abstract
Zika virus (ZIKV) is a clinically important flavivirus that can cause neurological disturbances in newborns. Here, we investigated comparatively the outcome of in vitro infection of newborn monocytes by ZIKV. We observed that neonatal cells show defective production of interleukin 1β, interleukin 10, and monocyte chemoattractant protein 1 in response to ZIKV, although they were as efficient as adult cells in supporting viral infection. Although CLEC5A is a classical flavivirus immune receptor, it is not essential to the cytokine response, but it regulates the viral load only in adult cells. Greater expression of viral entry receptors may create a favorable environment for viral invasion in neonatal monocytes. We are the first to suggest a role for CLEC5A in human monocyte infectivity and to show that newborn monocytes are interesting targets in ZIKV pathogenesis, owing to their ability to carry the virus with only a partial triggering of the immune response, creating a potentially favorable environment for virus-related pathologies in young individuals.
Collapse
Affiliation(s)
- Fabio Seiti Yamada Yoshikawa
- Laboratório de Investigação em Dermatologia e Imunodeficiências, Instituto de Medicina Tropical, Faculdade de Medicina
| | - Anna Julia Pietrobon
- Laboratório de Investigação em Dermatologia e Imunodeficiências, Instituto de Medicina Tropical, Faculdade de Medicina.,Departamento de Imunologia, Instituto de Ciências Biomédicas
| | - Anna Cláudia Calvielli Castelo Branco
- Laboratório de Investigação em Dermatologia e Imunodeficiências, Instituto de Medicina Tropical, Faculdade de Medicina.,Departamento de Imunologia, Instituto de Ciências Biomédicas
| | - Nátalli Zanete Pereira
- Laboratório de Investigação em Dermatologia e Imunodeficiências, Instituto de Medicina Tropical, Faculdade de Medicina.,Departamento de Imunologia, Instituto de Ciências Biomédicas
| | - Luanda Mara da Silva Oliveira
- Laboratório de Investigação em Dermatologia e Imunodeficiências, Instituto de Medicina Tropical, Faculdade de Medicina
| | | | - Alberto José da Silva Duarte
- Laboratório de Investigação em Dermatologia e Imunodeficiências, Instituto de Medicina Tropical, Faculdade de Medicina
| | - Maria Notomi Sato
- Laboratório de Investigação em Dermatologia e Imunodeficiências, Instituto de Medicina Tropical, Faculdade de Medicina.,Departamento de Imunologia, Instituto de Ciências Biomédicas
| |
Collapse
|
17
|
Abstract
The respiratory tract is tasked with responding to a constant and vast influx of foreign agents. It acts as an important first line of defense in the innate immune system and as such plays a crucial role in preventing the entry of invading pathogens. While physical barriers like the mucociliary escalator exert their effects through the clearance of these pathogens, diverse and dynamic cellular mechanisms exist for the activation of the innate immune response through the recognition of pathogen-associated molecular patterns (PAMPs). These PAMPs are recognized by pattern recognition receptors (PRRs) that are expressed on a number of myeloid cells such as dendritic cells, macrophages, and neutrophils found in the respiratory tract. C-type lectin receptors (CLRs) are PRRs that play a pivotal role in the innate immune response and its regulation to a variety of respiratory pathogens such as viruses, bacteria, and fungi. This chapter will describe the function of both activating and inhibiting myeloid CLRs in the recognition of a number of important respiratory pathogens as well as the signaling events initiated by these receptors.
Collapse
|
18
|
Sung PS, Hsieh SL. CLEC2 and CLEC5A: Pathogenic Host Factors in Acute Viral Infections. Front Immunol 2019; 10:2867. [PMID: 31867016 PMCID: PMC6909378 DOI: 10.3389/fimmu.2019.02867] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Accepted: 11/22/2019] [Indexed: 12/11/2022] Open
Abstract
The protective roles of endosomal toll-like receptors (TLRs) and cytosolic nucleic acid sensors are well elucidated, but the pathogenic host factors during viral infections remain unclear. Spleen tyrosine kinase (Syk)-coupled C-type lectins (CLECs) CLEC2 and CLEC5A are highly expressed on platelets and myeloid cells, respectively. CLEC2 has been shown to recognize snake venom aggretin and the endogenous ligand podoplanin and acts as a critical regulator in the development and immunothrombosis. Although CLEC2 has been reported to interact with type I immunodeficiency virus (HIV-1), its role in viral infections is still unclear. CLEC5A binds to fucose and mannose moieties of dengue virus membrane glycans, as well as to N-acetylglucosamine (GlcNAc)/N-acetylmuramic acid (MurNAc) disaccharides that form the backbone of L. monocytogenes peptidoglycans. Recently, we demonstrated that both CLEC2 and CLEC5A are critical in microbe-induced “neutrophil extracellular trap” (NET) formation and proinflammatory cytokine production. Moreover, activation of CLEC2 by dengue virus (DV) and H5N1 influenza virus (IAV) induces the release of extracellular vesicles (EVs), which further enhance NETosis and proinflammatory cytokine production via CLEC5A and Toll-like receptor 2 (TLR2). These findings not only illustrate the immunomodulatory effects of EVs during platelet-leukocyte interactions, but also demonstrate the critical roles of CLEC2 and CLEC5A in acute viral infections.
Collapse
Affiliation(s)
- Pei-Shan Sung
- Genomics Research Center, Academia Sinica, Taipei, Taiwan
| | - Shie-Liang Hsieh
- Genomics Research Center, Academia Sinica, Taipei, Taiwan.,Institute of Clinical Medicine, National Yang-Ming University, Taipei, Taiwan.,Department of Medical Research, Taipei Veterans General Hospital, Taipei, Taiwan.,Institute for Cancer Biology and Drug Discovery, Taipei Medical University, Taipei, Taiwan
| |
Collapse
|
19
|
Abstract
Neutrophils are implicated in almost every stage of oncogenesis and paradoxically display anti- and pro-tumor properties. Accumulating evidence indicates that neutrophils display diversity in their phenotype resulting from functional plasticity and/or changes to granulopoiesis. In cancer, neutrophils at a range of maturation stages can be identified in the blood and tissues (i.e., outside of their developmental niche). The functional capacity of neutrophils at different states of maturation is poorly understood resulting from challenges in their isolation, identification, and investigation. Thus, the impact of neutrophil maturity on cancer progression and therapy remains enigmatic. In this review, we discuss the identification, prevalence, and function of immature and mature neutrophils in cancer and the potential impact of this on tumor progression and cancer therapy.
Collapse
Affiliation(s)
- John B. G. Mackey
- Institute of Cancer Sciences, University of Glasgow, Glasgow, United Kingdom
- Cancer Research UK Beatson Institute, Glasgow, United Kingdom
| | - Seth B. Coffelt
- Institute of Cancer Sciences, University of Glasgow, Glasgow, United Kingdom
- Cancer Research UK Beatson Institute, Glasgow, United Kingdom
| | - Leo M. Carlin
- Institute of Cancer Sciences, University of Glasgow, Glasgow, United Kingdom
- Cancer Research UK Beatson Institute, Glasgow, United Kingdom
| |
Collapse
|
20
|
Schiffman C, McHale CM, Hubbard AE, Zhang L, Thomas R, Vermeulen R, Li G, Shen M, Rappaport SM, Yin S, Lan Q, Smith MT, Rothman N. Identification of gene expression predictors of occupational benzene exposure. PLoS One 2018; 13:e0205427. [PMID: 30300410 PMCID: PMC6177191 DOI: 10.1371/journal.pone.0205427] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2018] [Accepted: 09/25/2018] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Previously, using microarrays and mRNA-Sequencing (mRNA-Seq) we found that occupational exposure to a range of benzene levels perturbed gene expression in peripheral blood mononuclear cells. OBJECTIVES In the current study, we sought to identify gene expression biomarkers predictive of benzene exposure below 1 part per million (ppm), the occupational standard in the U.S. METHODS First, we used the nCounter platform to validate altered expression of 30 genes in 33 unexposed controls and 57 subjects exposed to benzene (<1 to ≥5 ppm). Second, we used SuperLearner (SL) to identify a minimal number of genes for which altered expression could predict <1 ppm benzene exposure, in 44 subjects with a mean air benzene level of 0.55±0.248 ppm (minimum 0.203ppm). RESULTS nCounter and microarray expression levels were highly correlated (coefficients >0.7, p<0.05) for 26 microarray-selected genes. nCounter and mRNA-Seq levels were poorly correlated for 4 mRNA-Seq-selected genes. Using negative binomial regression with adjustment for covariates and multiple testing, we confirmed differential expression of 23 microarray-selected genes in the entire benzene-exposed group, and 27 genes in the <1 ppm-exposed subgroup, compared with the control group. Using SL, we identified 3 pairs of genes that could predict <1 ppm benzene exposure with cross-validated AUC estimates >0.9 (p<0.0001) and were not predictive of other exposures (nickel, arsenic, smoking, stress). The predictive gene pairs are PRG2/CLEC5A, NFKBI/CLEC5A, and ACSL1/CLEC5A. They play roles in innate immunity and inflammatory responses. CONCLUSIONS Using nCounter and SL, we validated the altered expression of multiple mRNAs by benzene and identified gene pairs predictive of exposure to benzene at levels below the US occupational standard of 1ppm.
Collapse
Affiliation(s)
- Courtney Schiffman
- School of Public Health, University of California, Berkeley, California, United States of America
| | - Cliona M. McHale
- School of Public Health, University of California, Berkeley, California, United States of America
| | - Alan E. Hubbard
- School of Public Health, University of California, Berkeley, California, United States of America
| | - Luoping Zhang
- School of Public Health, University of California, Berkeley, California, United States of America
| | - Reuben Thomas
- School of Public Health, University of California, Berkeley, California, United States of America
| | - Roel Vermeulen
- Institute of Risk assessment Sciences, Utrecht University, Utrecht, the Netherlands
| | - Guilan Li
- Institute of Occupational Health and Poison Control, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Min Shen
- Occupational and Environmental Epidemiology Branch, Division of Cancer Epidemiology and Genetics, NCI, NIH, DHHS, Bethesda, Maryland, United States of America
| | - Stephen M. Rappaport
- School of Public Health, University of California, Berkeley, California, United States of America
| | - Songnian Yin
- Institute of Occupational Health and Poison Control, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Qing Lan
- Occupational and Environmental Epidemiology Branch, Division of Cancer Epidemiology and Genetics, NCI, NIH, DHHS, Bethesda, Maryland, United States of America
| | - Martyn T. Smith
- School of Public Health, University of California, Berkeley, California, United States of America
| | - Nathaniel Rothman
- Occupational and Environmental Epidemiology Branch, Division of Cancer Epidemiology and Genetics, NCI, NIH, DHHS, Bethesda, Maryland, United States of America
| |
Collapse
|
21
|
Molecular imaging assessment of periodontitis lesions in an experimental mouse model. Clin Oral Investig 2018; 23:821-827. [DOI: 10.1007/s00784-018-2510-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2017] [Accepted: 05/31/2018] [Indexed: 10/14/2022]
|
22
|
Ramezani A, Nahad MP, Faghihloo E. The role of Nrf2 transcription factor in viral infection. J Cell Biochem 2018; 119:6366-6382. [DOI: 10.1002/jcb.26897] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2017] [Accepted: 03/28/2018] [Indexed: 02/06/2023]
Affiliation(s)
- Ali Ramezani
- Virology DepartmentSchool of MedicineAhvaz Jundishapur University of Medical SciencesAhvazIran
- Hepatitis Research CenterBirjand University of Medical SciencesBirjandIran
| | - Mehdi Parsa Nahad
- Virology DepartmentSchool of MedicineAhvaz Jundishapur University of Medical SciencesAhvazIran
| | - Ebrahim Faghihloo
- Department of MicrobiologySchool of MedicineShahid Beheshti University of Medical SciencesTehranIran
| |
Collapse
|
23
|
Aoyagi H, Yamashiro K, Hirata‐Yoshihara C, Ideguchi H, Yamasaki M, Kawamura M, Yamamoto T, Kochi S, Wake H, Nishibori M, Takashiba S. HMGB1‐induced inflammatory response promotes bone healing in murine tooth extraction socket. J Cell Biochem 2018; 119:5481-5490. [DOI: 10.1002/jcb.26710] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2017] [Accepted: 01/23/2018] [Indexed: 12/26/2022]
Affiliation(s)
- Hiroaki Aoyagi
- Department of Pathophysiology—Periodontal ScienceOkayama University Graduate School of MedicineDentistry and Pharmaceutical SciencesOkayamaJapan
| | - Keisuke Yamashiro
- Department of Pathophysiology—Periodontal ScienceOkayama University Graduate School of MedicineDentistry and Pharmaceutical SciencesOkayamaJapan
| | - Chiaki Hirata‐Yoshihara
- Department of Pathophysiology—Periodontal ScienceOkayama University Graduate School of MedicineDentistry and Pharmaceutical SciencesOkayamaJapan
| | - Hidetaka Ideguchi
- Department of Pathophysiology—Periodontal ScienceOkayama University Graduate School of MedicineDentistry and Pharmaceutical SciencesOkayamaJapan
| | - Mutsuyo Yamasaki
- Department of Pathophysiology—Periodontal ScienceOkayama University Graduate School of MedicineDentistry and Pharmaceutical SciencesOkayamaJapan
| | - Mari Kawamura
- Department of Pathophysiology—Periodontal ScienceOkayama University Graduate School of MedicineDentistry and Pharmaceutical SciencesOkayamaJapan
| | - Tadashi Yamamoto
- Department of Pathophysiology—Periodontal ScienceOkayama University Graduate School of MedicineDentistry and Pharmaceutical SciencesOkayamaJapan
| | - Shinsuke Kochi
- Department of Pathophysiology—Periodontal ScienceOkayama University Graduate School of MedicineDentistry and Pharmaceutical SciencesOkayamaJapan
| | - Hidenori Wake
- Department of PharmacologyOkayama University Graduate School of MedicineDentistry and Pharmacological SciencesOkayamaJapan
| | - Masahiro Nishibori
- Department of PharmacologyOkayama University Graduate School of MedicineDentistry and Pharmacological SciencesOkayamaJapan
| | - Shogo Takashiba
- Department of Pathophysiology—Periodontal ScienceOkayama University Graduate School of MedicineDentistry and Pharmaceutical SciencesOkayamaJapan
| |
Collapse
|
24
|
Xavier-Carvalho C, Cardoso CC, de Souza Kehdy F, Pacheco AG, Moraes MO. Host genetics and dengue fever. INFECTION GENETICS AND EVOLUTION 2017; 56:99-110. [PMID: 29133029 DOI: 10.1016/j.meegid.2017.11.009] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/17/2017] [Revised: 11/07/2017] [Accepted: 11/09/2017] [Indexed: 12/13/2022]
Abstract
Dengue is a major worldwide problem in tropical and subtropical areas; it is caused by four different viral serotypes, and it can manifest as asymptomatic, mild, or severe. Many factors interact to determine the severity of the disease, including the genetic profile of the infected patient. However, the mechanisms that lead to severe disease and eventually death have not been determined, and a great challenge is the early identification of patients who are more likely to progress to a worse health condition. Studies performed in regions with cyclic outbreaks such as Cuba, Brazil, and Colombia have demonstrated that African ancestry confers protection against severe dengue. Highlighting the host genetics as an important factor in infectious diseases, a large number of association studies between genetic polymorphisms and dengue outcomes have been published in the last two decades. The most widely used approach involves case-control studies with candidate genes, such as the HLA locus and genes for receptors, cytokines, and other immune mediators. Additionally, a Genome-Wide Association Study (GWAS) identified SNPs associated with African ethnicity that had not previously been identified in case-control studies. Despite the increasing number of publications in America, Africa, and Asia, the results are quite controversial, and a meta-analysis is needed to assess the consensus among the studies. SNPs in the MICB, TNF, CD209, FcγRIIA, TPSAB1, CLEC5A, IL10 and PLCE1 genes are associated with the risk or protection of severe dengue, and the findings have been replicated in different populations. A thorough understanding of the viral, human genetic, and immunological mechanisms of dengue and how they interact is essential for effectively preventing dengue, but also managing and treating patients.
Collapse
Affiliation(s)
| | - Cynthia Chester Cardoso
- Laboratório de Virologia Molecular, Departamento de Genética, Instituto de Biologia, Universidade Federal do Rio de Janeiro, RJ, Brazil
| | | | | | - Milton Ozório Moraes
- Laboratório de Hanseníase, Instituto Oswaldo Cruz, FIOCRUZ, Rio de Janeiro, RJ, Brazil.
| |
Collapse
|
25
|
Xiong W, Wang H, Lu L, Xi R, Wang F, Gu G, Tao R. The macrophage C-type lectin receptor CLEC5A (MDL-1) expression is associated with early plaque progression and promotes macrophage survival. J Transl Med 2017; 15:234. [PMID: 29126450 PMCID: PMC5681784 DOI: 10.1186/s12967-017-1336-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2017] [Accepted: 10/30/2017] [Indexed: 12/11/2022] Open
Abstract
Background Biomarkers of early plaque progression are still elusive. Myeloid DAP12-associating lectin-1 (MDL-1), also called CLEC5A, is a C-type lectin receptor implicated in the progression of multiple acute and chronic inflammatory diseases. However, the relationship between its level and atherosclerosis is unknown. In this study, we aimed to investigate the correlation between macrophage MDL-1 expression and early atherosclerosis progression. Methods Immunofluorescence staining, real-time PCR and western blot were performed to analyze MDL-1 expression in aorta or mice macrophages. The role of MDL-1 in macrophage survival was further investigated by adenovirus infection and TUNEL assay. Results Significant MDL-1 expression was found in advanced human and apoE−/− mice atherosclerotic plaques, especially in lesional macrophages. In the model of atherosclerosis regression, we found MDL-1 expression was highly downregulated in lesional macrophages from ldlr−/− mouse regressive plaques, coincident with a reduction in lesional macrophage content and marker of M1 proinflammatory macrophages. Furthermore, we found MDL-1 was significantly expressed in inflammatory M1 subtype polarized bone marrow-derived macrophages. In vitro experiments, the level of MDL-1 was remarkably elevated in macrophages treated with pathophysiological drivers of plaque progression, such as oxidized low-density lipoprotein (ox-LDL) and hypoxia. Mechanistically, we demonstrated that MDL-1 overexpression notably promoted macrophage survival and decreased cleaved caspase-3 expression under ox-LDL stimulation, which suggested that it could maintain lesional macrophage survival and cause its accumulation. Conclusions This study firstly demonstrated that MDL-1 is mainly expressed in atherosclerotic lesional macrophages and increased macrophage MDL-1 expression is associated with early plaque progression and promotes macrophage survival. Electronic supplementary material The online version of this article (10.1186/s12967-017-1336-z) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Weixin Xiong
- Department of Cardiology, Ruijin Hospital, Jiao Tong University School of Medicine, No. 197, Ruijin Er Road, Huangpu District, Shanghai, 200025, China
| | - Haibo Wang
- Institute of Cardiovascular Disease, Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Lin Lu
- Department of Cardiology, Ruijin Hospital, Jiao Tong University School of Medicine, No. 197, Ruijin Er Road, Huangpu District, Shanghai, 200025, China.,Institute of Cardiovascular Disease, Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Rui Xi
- Department of Cardiology, Ruijin Hospital, Jiao Tong University School of Medicine, No. 197, Ruijin Er Road, Huangpu District, Shanghai, 200025, China
| | - Fang Wang
- Department of Cardiology, Ruijin Hospital, Jiao Tong University School of Medicine, No. 197, Ruijin Er Road, Huangpu District, Shanghai, 200025, China
| | - Gang Gu
- Department of Cardiology, Ruijin Hospital, Jiao Tong University School of Medicine, No. 197, Ruijin Er Road, Huangpu District, Shanghai, 200025, China.
| | - Rong Tao
- Department of Cardiology, Ruijin Hospital, Jiao Tong University School of Medicine, No. 197, Ruijin Er Road, Huangpu District, Shanghai, 200025, China.
| |
Collapse
|
26
|
Anti-Inflammatory Chromatinscape Suggests Alternative Mechanisms of Glucocorticoid Receptor Action. Immunity 2017; 47:298-309.e5. [PMID: 28801231 DOI: 10.1016/j.immuni.2017.07.012] [Citation(s) in RCA: 99] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2016] [Revised: 03/14/2017] [Accepted: 07/19/2017] [Indexed: 11/21/2022]
Abstract
Despite the widespread use of glucocorticoids (GCs), their anti-inflammatory effects are not understood mechanistically. Numerous investigations have examined the effects of glucocorticoid receptor (GR) activation prior to inflammatory challenges. However, clinical situations are emulated by a GC intervention initiated in the midst of rampant inflammatory responses. To characterize the effects of a late GC treatment, we profiled macrophage transcriptional and chromatinscapes with Dexamethasone (Dex) treatment before or after stimulation by lipopolysaccharide (LPS). The late activation of GR had a similar gene-expression profile as from GR pre-activation, while ameliorating the disruption of metabolic genes. Chromatin occupancy of GR was not predictive of Dex-regulated gene expression, contradicting the "trans-repression by tethering" model. Rather, GR activation resulted in genome-wide blockade of NF-κB interaction with chromatin and directly induced inhibitors of NF-κB and AP-1. Our investigation using GC treatments with clinically relevant timing highlights mechanisms underlying GR actions for modulating the "inflamed epigenome."
Collapse
|
27
|
Xavier-Carvalho C, Cezar RDDS, Freire NM, Vasconcelos CMMD, Solorzano VEF, de Toledo-Pinto TG, Fialho LG, do Carmo RF, Vasconcelos LRS, Cordeiro MT, Baptista P, de Azeredo EL, da Cunha RV, de Souza LJ, Pacheco AG, Kubelka CF, Moura PMMFD, Moraes MO. Association of rs1285933 single nucleotide polymorphism in CLEC5A gene with dengue severity and its functional effects. Hum Immunol 2017; 78:649-656. [PMID: 28764923 DOI: 10.1016/j.humimm.2017.07.013] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2016] [Revised: 06/26/2017] [Accepted: 07/27/2017] [Indexed: 12/16/2022]
Abstract
Outbreaks of the Zika, dengue, and chikungunya viruses, especially in the Americas, pose a global threat due to their rapid spread and difficulty controlling the vector. Extreme phenotypes are often observed, from asymptomatic to severe clinical manifestations, which are well-studied in dengue. Host variations are also important contributors to disease outcomes, and many case-control studies have associated single nucleotide polymorphisms (SNPs) with severe dengue. Here, we found that the TC genotype and T-carriers for SNP rs1285933 in the C-type lectin superfamily member 5 (CLEC5A) gene was associated with severe dengue in a Northern Brazilian population (OR=2.75 and p-value=0.01, OR=2.11 and p-value=0.04, respectively). We also tested the functional effect of the CLEC5A protein and found that it is upregulated on the surface of human monocytes after in vitro dengue infection. CLEC5A was correlated with viral load inside the monocytes (Spearman r=0.55, p=0.008) and TNF production in culture supernatants (Spearman r=0.72, p=0.03). Analysis of mRNA in blood samples from DENV4-infected patients exhibiting mild symptoms showed that CLEC5A mRNA expression is correlated with TNF (r=0.67, p=0.0001) and other immune mediators. Monocytes from rs1285933 TT/TC individuals showed lower CLEC5A expression compared to CC genotypes. However, in these cells, CLEC5A was not correlated with TNF production. In summary, we confirmed that CLEC5A is genetically associated with dengue severity outcome, playing a central role during the immune response triggered by a dengue viral infection, and rs1285933 is a relevant SNP that is able to regulate signaling pathways after interactions between the dengue virus and CLEC5A receptors.
Collapse
Affiliation(s)
- Caroline Xavier-Carvalho
- Laboratório de Hanseníase, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, FIOCRUZ, Rio de Janeiro, RJ, Brazil; Laboratório de Imunologia Viral, Instituto Oswaldo Cruz - FIOCRUZ, Rio de Janeiro, RJ, Brazil
| | | | - Naishe Matos Freire
- Faculdade de Ciências Médicas, Universidade de Pernambuco, Recife, PE, Brazil
| | | | | | | | - Luciana Gomes Fialho
- Laboratório de Imunologia Viral, Instituto Oswaldo Cruz - FIOCRUZ, Rio de Janeiro, RJ, Brazil
| | - Rodrigo Feliciano do Carmo
- Colegiado de Ciências Farmacêuticas, Universidade Federal do Vale do São Francisco, Petrolina, PE, Brazil
| | - Luydson Richardson Silva Vasconcelos
- Faculdade de Ciências Médicas, Universidade de Pernambuco, Recife, PE, Brazil; Laboratório de Doenças Transmissíveis, Departamento de Parasitologia, FIOCRUZ, Recife, PE, Brazil
| | | | - Paulo Baptista
- Faculdade de Ciências Médicas, Universidade de Pernambuco, Recife, PE, Brazil
| | | | - Rivaldo Venâncio da Cunha
- Departamento de Clínica Médica, FM, Universidade Federal do Mato Grosso do Sul, Campo Grande, MS, Brazil
| | - Luiz José de Souza
- Centro de Referência em Dengue e Faculdade de Medicina, Campos de Goytacazes, RJ, Brazil
| | | | | | - Patrícia Muniz Mendes Freire de Moura
- Instituto de Ciências Biológicas, Universidade de Pernambuco, Recife, PE, Brazil; Laboratório de Doenças Transmissíveis, Departamento de Parasitologia, FIOCRUZ, Recife, PE, Brazil
| | - Milton Ozorio Moraes
- Laboratório de Hanseníase, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, FIOCRUZ, Rio de Janeiro, RJ, Brazil.
| |
Collapse
|
28
|
Sprokholt J, Helgers LC, Geijtenbeek TBH. Innate immune receptors drive dengue virus immune activation and disease. Future Virol 2017; 13:287-305. [PMID: 29937918 PMCID: PMC6004600 DOI: 10.2217/fvl-2017-0146] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2017] [Accepted: 01/25/2018] [Indexed: 12/14/2022]
Abstract
Dengue is a worldwide disease with 400 million annual infections that can lead to septic shock and viral hemorrhagic fever with internal bleeding. These symptoms are the result of uncontrolled immune activation. Macrophages and dendritic cells are the main target of dengue virus (DENV) and the cellular source of cytokines associated with this immune activation. Macrophages and dendritic cells express several innate immune receptors that have been implicated in DENV immune activation, of which, CLEC5A, RIG-I and MDA5 are most important. Notably, activation of these receptors have profound effects on adaptive immune responses against DENV. This review will focus on how innate immune receptors drive DENV immune activation by inducing inflammatory cytokines and by activating adaptive immune responses.
Collapse
Affiliation(s)
- Joris Sprokholt
- Department of Experimental Immunology, Academic Medical Center, University of Amsterdam, 1105AZ Amsterdam, The Netherlands
- Amsterdam Infection & Immunity Institute, AMC, VUmc, Amsterdam, The Netherlands
| | - Leanne C Helgers
- Department of Experimental Immunology, Academic Medical Center, University of Amsterdam, 1105AZ Amsterdam, The Netherlands
- Amsterdam Infection & Immunity Institute, AMC, VUmc, Amsterdam, The Netherlands
| | - Teunis BH Geijtenbeek
- Department of Experimental Immunology, Academic Medical Center, University of Amsterdam, 1105AZ Amsterdam, The Netherlands
- Amsterdam Infection & Immunity Institute, AMC, VUmc, Amsterdam, The Netherlands
| |
Collapse
|
29
|
FTY720 Attenuates Angiotensin II-Induced Podocyte Damage via Inhibiting Inflammatory Cytokines. Mediators Inflamm 2017; 2017:3701385. [PMID: 28270699 PMCID: PMC5320072 DOI: 10.1155/2017/3701385] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2016] [Revised: 12/08/2016] [Accepted: 12/26/2016] [Indexed: 12/29/2022] Open
Abstract
FTY720, a new chemical substance derived from the ascomycete Isaria sinclairii, is used for treating multiple sclerosis, renal cancer, and asthma. Sphingosine 1-phosphate (S1P) is a bioactive sphingolipid metabolite and exists in red blood cells. FTY720 is a synthetic S1P analog which can block S1P evoking physiological effects. Recently studies show that S1P was participating in activated inflammation cells induced renal injury. The objective of this study was to assess the protective effect of FTY720 on kidney damage and the potential mechanism of FTY720 which alleviate podocyte injury in chronic kidney disease. In this study, we selected 40 patients with IgA nephropathy and examined their clinical characteristics. Ang II-infusion rat renal injury model was established to evaluate the glomeruli and tubulointerstitial lesion. The result showed that the concentration of S1P in serum and urine was positively correlated with IgA nephropathy patients' renal injury. FTY720 could reduce renal histological lesions induced by Ang II-infusion in rats. Moreover, FTY720 decreased S1P synthesis in Ang II-infusion rats via downregulation of inflammatory cytokines including TNF-α and IL-6. In addition, FTY720 alleviated exogenous S1P-induced podocyte damage. In conclusion, FTY720 is able to attenuate S1P-induced podocyte damage via reducing inflammatory cytokines.
Collapse
|
30
|
CLEC5A-Mediated Enhancement of the Inflammatory Response in Myeloid Cells Contributes to Influenza Virus Pathogenicity In Vivo. J Virol 2016; 91:JVI.01813-16. [PMID: 27795434 PMCID: PMC5165214 DOI: 10.1128/jvi.01813-16] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2016] [Accepted: 10/14/2016] [Indexed: 12/27/2022] Open
Abstract
Human infections with influenza viruses exhibit mild to severe clinical outcomes as a result of complex virus-host interactions. Induction of inflammatory mediators via pattern recognition receptors may dictate subsequent host responses for pathogen clearance and tissue damage. We identified that human C-type lectin domain family 5 member A (CLEC5A) interacts with the hemagglutinin protein of influenza viruses expressed on lentiviral pseudoparticles through lectin screening. Silencing CLEC5A gene expression, blocking influenza-CLEC5A interactions with anti-CLEC5A antibodies, or dampening CLEC5A-mediated signaling using a spleen tyrosine kinase inhibitor consistently reduced the levels of proinflammatory cytokines produced by human macrophages without affecting the replication of influenza A viruses of different subtypes. Infection of bone marrow-derived macrophages from CLEC5A-deficient mice showed reduced levels of tumor necrosis factor alpha (TNF-α) and IP-10 but elevated alpha interferon (IFN-α) compared to those of wild-type mice. The heightened type I IFN response in the macrophages of CLEC5A-deficient mice was associated with upregulated TLR3 mRNA after treatment with double-stranded RNA. Upon lethal challenges with a recombinant H5N1 virus, CLEC5A-deficient mice showed reduced levels of proinflammatory cytokines, decreased immune cell infiltration in the lungs, and improved survival compared to the wild-type mice, despite comparable viral loads noted throughout the course of infection. The survival difference was more prominent at a lower dose of inoculum. Our results suggest that CLEC5A-mediated enhancement of the inflammatory response in myeloid cells contributes to influenza pathogenicity in vivo and may be considered a therapeutic target in combination with effective antivirals. Well-orchestrated host responses together with effective viral clearance are critical for optimal clinical outcome after influenza infections.
IMPORTANCE Multiple pattern recognition receptors work in synergy to sense viral RNA or proteins synthesized during influenza replication and mediate host responses for viral control. Well-orchestrated host responses may help to maintain the inflammatory response to minimize tissue damage while inducing an effective adaptive immune response for viral clearance. We identified that CLEC5A, a C-type lectin receptor which has previously been reported to mediate flavivirus-induced inflammatory responses, enhanced induction of proinflammatory cytokines and chemokines in myeloid cells after influenza infections. CLEC5A-deficient mice infected with influenza virus showed reduced inflammation in the lungs and improved survival compared to that of the wild-type mice despite comparable viral loads. The survival difference was more prominent at a lower dose of inoculum. Collectively, our results suggest that dampening CLEC5A-mediated inflammatory responses in myeloid cells reduces immunopathogenesis after influenza infections.
Collapse
|
31
|
Dengue Virus Infection Is through a Cooperative Interaction between a Mannose Receptor and CLEC5A on Macrophage as a Multivalent Hetero-Complex. PLoS One 2016; 11:e0166474. [PMID: 27832191 PMCID: PMC5104462 DOI: 10.1371/journal.pone.0166474] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2016] [Accepted: 10/28/2016] [Indexed: 01/01/2023] Open
Abstract
Dengue fever is a mosquito-borne viral pandemic disease that is widespread in the tropical and subtropical areas. Dengue virus uses human mannose-binding receptor (MR) and DC-SIGN on macrophages as primary receptors, and CLEC5A as signaling receptor to sense the dengue virus invasion and then to signal and stimulate macrophages to secrete cytokines. But the interplay between MR/DC-SIGN and CLEC5A is unknown. Here we demonstrate a plausible mechanism for the interaction, i.e. MR/DC-SIGN first attracts the virus with high avidity, and the virus concurrently interacts with CLEC5A in close proximity to form a multivalent hetero-complex and facilitate CLEC5A-mediated signal transduction. Our study suggests that the cooperation between a high-avidity lectin-virus interaction and a nearby low-avidity signaling receptor provides a necessary connection between binding and signaling. Understanding this mechanism may lead to the development of a new antiviral strategy.
Collapse
|
32
|
Yost CC, Schwertz H, Cody MJ, Wallace JA, Campbell RA, Vieira-de-Abreu A, Araujo CV, Schubert S, Harris ES, Rowley JW, Rondina MT, Fulcher JM, Koening CL, Weyrich AS, Zimmerman GA. Neonatal NET-inhibitory factor and related peptides inhibit neutrophil extracellular trap formation. J Clin Invest 2016; 126:3783-3798. [PMID: 27599294 DOI: 10.1172/jci83873] [Citation(s) in RCA: 111] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2015] [Accepted: 07/28/2016] [Indexed: 12/22/2022] Open
Abstract
Neutrophil granulocytes, also called polymorphonuclear leukocytes (PMNs), extrude molecular lattices of decondensed chromatin studded with histones, granule enzymes, and antimicrobial peptides that are referred to as neutrophil extracellular traps (NETs). NETs capture and contain bacteria, viruses, and other pathogens. Nevertheless, experimental evidence indicates that NETs also cause inflammatory vascular and tissue damage, suggesting that identifying pathways that inhibit NET formation may have therapeutic implications. Here, we determined that neonatal NET-inhibitory factor (nNIF) is an inhibitor of NET formation in umbilical cord blood. In human neonatal and adult neutrophils, nNIF inhibits key terminal events in NET formation, including peptidyl arginine deiminase 4 (PAD4) activity, neutrophil nuclear histone citrullination, and nuclear decondensation. We also identified additional nNIF-related peptides (NRPs) that inhibit NET formation. nNIFs and NRPs blocked NET formation induced by pathogens, microbial toxins, and pharmacologic agonists in vitro and in mouse models of infection and systemic inflammation, and they improved mortality in murine models of systemic inflammation, which are associated with NET-induced collateral tissue injury. The identification of NRPs as neutrophil modulators that selectively interrupt NET generation at critical steps suggests their potential as therapeutic agents. Furthermore, our results indicate that nNIF may be an important regulator of NET formation in fetal and neonatal inflammation.
Collapse
|
33
|
Cheng YL, Lin YS, Chen CL, Tsai TT, Tsai CC, Wu YW, Ou YD, Chu YY, Wang JM, Yu CY, Lin CF. Activation of Nrf2 by the dengue virus causes an increase in CLEC5A, which enhances TNF-α production by mononuclear phagocytes. Sci Rep 2016; 6:32000. [PMID: 27561946 PMCID: PMC4999957 DOI: 10.1038/srep32000] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2016] [Accepted: 07/27/2016] [Indexed: 12/11/2022] Open
Abstract
Infection by the dengue virus (DENV) threatens global public health due to its high prevalence and the lack of effective treatments. Host factors may contribute to the pathogenesis of DENV; herein, we investigated the role of nuclear factor (erythroid-derived 2)-like 2 (Nrf2), which is activated by DENV in mononuclear phagocytes. DENV infection selectively activates Nrf2 following nuclear translocation. Following endoplasmic reticular (ER) stress, protein kinase R-like ER kinase (PERK) facilitated Nrf2-mediated transcriptional activation of C-type lectin domain family 5, member A (CLEC5A) to increase CLEC5A expression. Signaling downstream of the Nrf2-CLEC5A interaction enhances Toll-like receptor 3 (TLR3)-independent tumor necrosis factor (TNF)-α production following DENV infection. Forced expression of the NS2B3 viral protein induces Nrf2 nuclear translocation/activation and CLEC5A expression which increases DENV-induced TNF-α production. Animal studies confirmed Nrf2-induced CLEC5A and TNF-α in brains of DENV-infected mice. These results demonstrate that DENV infection causes Nrf2-regulated TNF-α production by increasing levels of CLEC5A.
Collapse
Affiliation(s)
- Yi-Lin Cheng
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan 701, Taiwan.,Center of Infectious Disease and Signaling Research, National Cheng Kung University, Tainan 701, Taiwan
| | - Yee-Shin Lin
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan 701, Taiwan.,Center of Infectious Disease and Signaling Research, National Cheng Kung University, Tainan 701, Taiwan.,Department of Microbiology and Immunology, College of Medicine, National Cheng Kung University, Tainan 701, Taiwan
| | - Chia-Ling Chen
- Translational Research Center, Taipei Medical University, Taipei 110, Taiwan
| | - Tsung-Ting Tsai
- Department of Microbiology and Immunology, College of Medicine, Taipei Medical University, Taipei 110, Taiwan
| | - Cheng-Chieh Tsai
- Department of Nursing, Chung Hwa University of Medical Technology, Tainan 717, Taiwan
| | - Yan-Wei Wu
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan 701, Taiwan.,Center of Infectious Disease and Signaling Research, National Cheng Kung University, Tainan 701, Taiwan
| | - Yi-Dan Ou
- Department of Microbiology and Immunology, College of Medicine, National Cheng Kung University, Tainan 701, Taiwan
| | - Yu-Yi Chu
- Institute of Bioinformatics and Biosignal Transduction, National Cheng Kung University, Tainan 701, Taiwan
| | - Ju-Ming Wang
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan 701, Taiwan.,Center of Infectious Disease and Signaling Research, National Cheng Kung University, Tainan 701, Taiwan.,Institute of Bioinformatics and Biosignal Transduction, National Cheng Kung University, Tainan 701, Taiwan
| | - Chia-Yi Yu
- Department of Microbiology and Immunology, College of Medicine, National Cheng Kung University, Tainan 701, Taiwan
| | - Chiou-Feng Lin
- Center of Infectious Disease and Signaling Research, National Cheng Kung University, Tainan 701, Taiwan.,Department of Microbiology and Immunology, College of Medicine, Taipei Medical University, Taipei 110, Taiwan.,Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei 110, Taiwan
| |
Collapse
|
34
|
Heinrich A, Heyl KA, Klaile E, Müller MM, Klassert TE, Wiessner A, Fischer K, Schumann RR, Seifert U, Riesbeck K, Moter A, Singer BB, Bachmann S, Slevogt H. Moraxella catarrhalis induces CEACAM3-Syk-CARD9-dependent activation of human granulocytes. Cell Microbiol 2016; 18:1570-1582. [PMID: 27038042 PMCID: PMC5096018 DOI: 10.1111/cmi.12597] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2014] [Revised: 03/18/2016] [Accepted: 03/21/2016] [Indexed: 12/14/2022]
Abstract
The human restricted pathogen Moraxella catarrhalis is an important causal agent for exacerbations in chronic obstructive lung disease in adults. In such patients, increased numbers of granulocytes are present in the airways, which correlate with bacteria-induced exacerbations and severity of the disease. Our study investigated whether the interaction of M. catarrhalis with the human granulocyte-specific carcinoembryonic antigen-related cell adhesion molecule (CEACAM)-3 is linked to NF-κB activation, resulting in chemokine production. Granulocytes from healthy donors and NB4 cells were infected with M. catarrhalis in the presence of different inhibitors, blocking antibodies and siRNA. The supernatants were analysed by enzyme-linked immunosorbent assay for chemokines. NF-κB activation was determined using a luciferase reporter gene assay and chromatin-immunoprecipitation. We found evidence that the specific engagement of CEACAM3 by M. catarrhalis ubiquitous surface protein A1 (UspA1) results in the activation of pro-inflammatory events, such as degranulation of neutrophils, ROS production and chemokine secretion. The interaction of UspA1 with CEACAM3 induced the activation of the NF-κB pathway via Syk and the CARD9 pathway and was dependent on the phosphorylation of the CEACAM3 ITAM-like motif. These findings suggest that the CEACAM3 signalling in neutrophils is able to specifically modulate airway inflammation caused by infection with M. catarrhalis.
Collapse
Affiliation(s)
- A Heinrich
- Septomics Research Center, Jena University Hospital, Jena, Germany
| | - K A Heyl
- Septomics Research Center, Jena University Hospital, Jena, Germany
| | - E Klaile
- Septomics Research Center, Jena University Hospital, Jena, Germany.,Center for Sepsis Control and Care, Jena University Hospital, Jena, Germany
| | - M M Müller
- Septomics Research Center, Jena University Hospital, Jena, Germany
| | - T E Klassert
- Septomics Research Center, Jena University Hospital, Jena, Germany
| | - A Wiessner
- Biofilmcenter, German Heart Institute Berlin, Berlin, Germany
| | - K Fischer
- Septomics Research Center, Jena University Hospital, Jena, Germany.,Department of Infectious Diseases and Pulmonary Medicine, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - R R Schumann
- Institute for Microbiology and Hygiene, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - U Seifert
- Institute of Molecular and Clinical Immunology, Medical Faculty, Otto-von-Guericke University, Magdeburg, Germany
| | - K Riesbeck
- Clinical Microbiology, Department of Translational Medicine, Lund University, Malmö, Sweden
| | - A Moter
- Biofilmcenter, German Heart Institute Berlin, Berlin, Germany
| | - B B Singer
- Institute of Anatomy, University Hospital, University Duisburg-Essen, Essen, Germany
| | - S Bachmann
- Institute of Vegetative Anatomy, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - H Slevogt
- Septomics Research Center, Jena University Hospital, Jena, Germany.
| |
Collapse
|
35
|
Wortham BW, Eppert BL, Flury JL, Garcia SM, Donica WR, Osterburg A, Joyce-Shaikh B, Cua DJ, Borchers MT. Cutting Edge: CLEC5A Mediates Macrophage Function and Chronic Obstructive Pulmonary Disease Pathologies. THE JOURNAL OF IMMUNOLOGY 2016; 196:3227-31. [PMID: 26927798 DOI: 10.4049/jimmunol.1500978] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/01/2015] [Accepted: 02/12/2016] [Indexed: 12/21/2022]
Abstract
Chronic obstructive pulmonary disease (COPD) is a devastating disease with no effective therapies. We investigated the role of the C-type lectin receptor, CLEC5A, in macrophage activation and pulmonary pathogenesis in a mouse model of COPD. We demonstrate that CLEC5A is expressed on alveolar macrophages in mice exposed long-term to cigarette smoke (CS), as well as in human smokers. We also show that CLEC5A-mediated activation of macrophages enhanced cytokine elaboration alone, as well as in combination with LPS or GM-CSF in CS-exposed mice. Furthermore, usingClec5a-deficient mice, we demonstrate that CS-induced macrophage responsiveness is mediated by CLEC5A, and CLEC5A is required for the development of inflammation, proinflammatory cytokine expression, and airspace enlargement. These findings suggest a novel mechanism that promotes airway inflammation and pathologies in response to CS exposure and identifies CLEC5A as a novel target for the therapeutic control of COPD pathogenesis.
Collapse
Affiliation(s)
- Brian W Wortham
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, University of Cincinnati College of Medicine, Cincinnati, OH 45267; and
| | - Bryan L Eppert
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, University of Cincinnati College of Medicine, Cincinnati, OH 45267; and
| | - Jennifer L Flury
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, University of Cincinnati College of Medicine, Cincinnati, OH 45267; and
| | - Sara Morgado Garcia
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, University of Cincinnati College of Medicine, Cincinnati, OH 45267; and
| | - Walter R Donica
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, University of Cincinnati College of Medicine, Cincinnati, OH 45267; and
| | - Andrew Osterburg
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, University of Cincinnati College of Medicine, Cincinnati, OH 45267; and
| | | | - Daniel J Cua
- Merck Research Laboratories, Palo Alto, CA 94304
| | - Michael T Borchers
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, University of Cincinnati College of Medicine, Cincinnati, OH 45267; and
| |
Collapse
|
36
|
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
|
37
|
Chen X, Eksioglu EA, Carter JD, Fortenbery N, Donatelli SS, Zhou J, Liu J, Yang L, Gilvary D, Djeu J, Wei S. Inactivation of DAP12 in PMN inhibits TREM1-mediated activation in rheumatoid arthritis. PLoS One 2015; 10:e0115116. [PMID: 25642940 PMCID: PMC4313943 DOI: 10.1371/journal.pone.0115116] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2014] [Accepted: 11/19/2014] [Indexed: 11/18/2022] Open
Abstract
Rheumatoid arthritis (RA) is an autoimmune disease characterized by dysregulated and chronic systemic inflammatory responses that affect the synovium, bone, and cartilage causing damage to extra-articular tissue. Innate immunity is the first line of defense against invading pathogens and assists in the initiation of adaptive immune responses. Polymorphonuclear cells (PMNs), which include neutrophils, are the largest population of white blood cells in peripheral blood and functionally produce their inflammatory effect through phagocytosis, cytokine production and natural killer-like cytotoxic activity. TREM1 (triggering receptor expressed by myeloid cells) is an inflammatory receptor in PMNs that signals through the use of the intracellular activating adaptor DAP12 to induce downstream signaling. After TREM crosslinking, DAP12's tyrosines in its ITAM motif get phosphorylated inducing the recruitment of Syk tyrosine kinases and eventual activation of PI3 kinases and ERK signaling pathways. While both TREM1 and DAP12 have been shown to be important activators of RA pathogenesis, their activity in PMNs or the importance of DAP12 as a possible therapeutic target have not been shown. Here we corroborate, using primary RA specimens, that isolated PMNs have an increased proportion of both TREM1 and DAP12 compared to normal healthy control isolated PMNs both at the protein and gene expression levels. This increased expression is highly functional with increased activation of ERK and MAPKs, secretion of IL-8 and RANTES and cytotoxicity of target cells. Importantly, based on our hypothesis of an imbalance of activating and inhibitory signaling in the pathogenesis of RA we demonstrate that inhibition of the DAP12 signaling pathway inactivates these important inflammatory cells.
Collapse
Affiliation(s)
- Xianghong Chen
- Department of Immunology, H. Lee Moffitt Cancer Center, Tampa, Florida, United States of America
| | - Erika A. Eksioglu
- Department of Immunology, H. Lee Moffitt Cancer Center, Tampa, Florida, United States of America
| | - John D. Carter
- Department of Internal Medicine, Division of Rheumatology, University of South Florida College of Medicine, Tampa, Florida, United States of America
| | - Nicole Fortenbery
- Department of Immunology, H. Lee Moffitt Cancer Center, Tampa, Florida, United States of America
| | - Sarah S. Donatelli
- Department of Immunology, H. Lee Moffitt Cancer Center, Tampa, Florida, United States of America
| | - Junmin Zhou
- Department of Immunology, H. Lee Moffitt Cancer Center, Tampa, Florida, United States of America
| | - Jinhong Liu
- Department of Immunology, H. Lee Moffitt Cancer Center, Tampa, Florida, United States of America
| | - Lili Yang
- Department of Immunology, Tianjin Medical University Cancer Hospital and Research Institute, Tianjin, China
| | - Danielle Gilvary
- Department of Immunology, H. Lee Moffitt Cancer Center, Tampa, Florida, United States of America
| | - Julie Djeu
- Department of Immunology, H. Lee Moffitt Cancer Center, Tampa, Florida, United States of America
| | - Sheng Wei
- Department of Immunology, H. Lee Moffitt Cancer Center, Tampa, Florida, United States of America
- Department of Immunology, Tianjin Medical University Cancer Hospital and Research Institute, Tianjin, China
- * E-mail:
| |
Collapse
|
38
|
SSC(high)CD11b(high)Ly-6C(high)Ly-6G(low) myeloid cells curtail CD4 T cell response by inducible nitric oxide synthase in murine hepatitis. Int J Biochem Cell Biol 2014; 54:89-97. [PMID: 25035167 DOI: 10.1016/j.biocel.2014.07.005] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2014] [Revised: 04/28/2014] [Accepted: 07/04/2014] [Indexed: 12/16/2022]
Abstract
Myeloid-derived suppressor cells (MDSCs) play an important role in maintaining immune tolerance in response to tumors and inflammatory diseases. Several liver MDSCs have been described in hepatitis in humans and mouse models. Although all the murine MDSCs are CD11b(+)Gr-1(+), their true phenotype and mechanism of suppression remain elusive. This study revealed that SSC(high)CD11b(high)Ly-6C(high)Ly-6G(low) monocytic cells but not the other liver-infiltrating, CD11b(+)Gr-1(+) subsets could suppress CD4 T cell responses. Their suppressive activity was remarkably effective even at a ratio of 1:50 when co-cultured with CD4 T cells. Mechanistically, the suppression was dependent on nitric oxide production by inducible nitric oxide synthase (iNOS). Furthermore, the suppressive function by these liver MDSCs was found to require direct contact with activated CD4 T cells. Adoptive transfer experiments demonstrate that these liver MDSCs can dramatically ameliorate concanavalin A (Con A)-induced fulminant hepatitis in mice. Finally, MDSC-mediated suppression in vivo was dependent on iNOS expression. Altogether, SSC(high)CD11b(high)Ly-6C(high)Ly-6G(low) cells represent authentic MDSCs in the inflammatory liver and may function to minimize collateral damage caused by an overzealous CD4 T cell response following hepatitis infection.
Collapse
|
39
|
Elpek KG, Cremasco V, Shen H, Harvey CJ, Wucherpfennig KW, Goldstein DR, Monach PA, Turley SJ. The tumor microenvironment shapes lineage, transcriptional, and functional diversity of infiltrating myeloid cells. Cancer Immunol Res 2014; 2:655-67. [PMID: 24801837 DOI: 10.1158/2326-6066.cir-13-0209] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Myeloid cells play important regulatory roles within the tumor environment by directly promoting tumor progression and modulating the function of tumor-infiltrating lymphocytes, and as such, they represent a potential therapeutic target for the treatment of cancer. Although distinct subsets of tumor-associated myeloid cells have been identified, a broader analysis of the complete myeloid cell landscape within individual tumors and also across different tumor types has been lacking. By establishing the developmental and transcriptomic signatures of infiltrating myeloid cells from multiple primary tumors, we found that tumor-associated macrophages (TAM) and tumor-associated neutrophils (TAN), while present within all tumors analyzed, exhibited strikingly different frequencies, gene expression profiles, and functions across cancer types. We also evaluated the impact of anatomic location and circulating factors on the myeloid cell composition of tumors. The makeup of the myeloid compartment was determined by the tumor microenvironment rather than the anatomic location of tumor development or tumor-derived circulating factors. Protumorigenic and hypoxia-associated genes were enriched in TAMs and TANs compared with splenic myeloid-derived suppressor cells. Although all TANs had an altered expression pattern of secretory effector molecules, in each tumor type they exhibited a unique cytokine, chemokine, and associated receptor expression profile. One such molecule, haptoglobin, was uniquely expressed by 4T1 TANs and identified as a possible diagnostic biomarker for tumors characterized by the accumulation of myeloid cells. Thus, we have identified considerable cancer-specific diversity in the lineage, gene expression, and function of tumor-infiltrating myeloid cells.
Collapse
Affiliation(s)
- Kutlu G Elpek
- Authors' Affiliations: Department of Cancer Immunology and AIDS, Dana-Farber Cancer Institute
| | - Viviana Cremasco
- Authors' Affiliations: Department of Cancer Immunology and AIDS, Dana-Farber Cancer Institute
| | - Hua Shen
- Internal Medicine and Immunobiology, Yale School of Medicine, New Haven, Connecticut
| | - Christopher J Harvey
- Authors' Affiliations: Department of Cancer Immunology and AIDS, Dana-Farber Cancer Institute
| | - Kai W Wucherpfennig
- Authors' Affiliations: Department of Cancer Immunology and AIDS, Dana-Farber Cancer Institute
| | - Daniel R Goldstein
- Internal Medicine and Immunobiology, Yale School of Medicine, New Haven, Connecticut
| | | | - Shannon J Turley
- Authors' Affiliations: Department of Cancer Immunology and AIDS, Dana-Farber Cancer Institute; Department of Microbiology and Immunobiology, Harvard Medical School, Boston, Massachusetts; Departments of
| |
Collapse
|
40
|
Yin XD, Jia PJ, Pang Y, He JH. Protective effect of FTY720 on several markers of liver injury induced by concanavalin a in mice. Curr Ther Res Clin Exp 2014; 73:140-9. [PMID: 24653516 DOI: 10.1016/j.curtheres.2012.07.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/26/2012] [Indexed: 10/27/2022] Open
Abstract
BACKGROUND 2-Amino-2-[2-(4-octylphenyl)ethyl] propane-1,3-diol hydrochloride (FTY720) is a novel agent with protective effect on several markers of liver injury. It is a chemical substance derived by modifying myriocin from the ascomycete Isaria sinclairii. It has been reported that FTY720 is able to treat autoimmune encephalomyelitis, renal cancer, asthma, and multiple sclerosis. More potent clinical applications of FTY720 need to be investigated. OBJECTIVE The aim of this study was to evaluate the protective effect of FTY720 on several markers of experimental liver injury and to investigate the possible mechanism of action. METHODS Concanavalin A (Con A) at a dose of 15 mg/kg was intravenously. injected in mice, and 10 days before the Con A challenge, 1 mg/kg, 3 mg/kg, and 6 mg/kg of FTY720 were administered to mice. The liver injury was monitored biochemically by measuring serum alanine aminotransferase (ALT) and aspartate aminotransferase (AST) and tumor necrosis factor-α (TNF-α) levels. TNF-α and nuclear factor-κB (NF-κB) in liver tissue were detected by Western blot analysis. RESULTS FTY720, when administered intragastrically for 10 days in mice with Con A-induced liver injury, dose-dependently reduced serum ALT and AST and TNF-α levels. The differences were statistically significant (P ≤ 0.05). It was also found that FTY720 decreases TNF-α and NF-κB protein expression in liver tissue. CONCLUSIONS FTY720 is able to improve several markers of Con A-induced liver injury in mice, including serum ALT, serum AST, TNF-α, and NF-κB, which might be at least in part related to its ability to reduce TNF-α/NF-κB cascade activity.
Collapse
Affiliation(s)
- Xiao-Dong Yin
- Department of Oncology, Tianjin Union Medicine Center, Tianjin, China
| | - Pei-Jie Jia
- Department of Oncology, Tianjin Union Medicine Center, Tianjin, China
| | - Yan Pang
- Department of Oncology, Tianjin Union Medicine Center, Tianjin, China
| | - Jing-Hua He
- Department of Pharmacology, Basic Medical School, Tianjin Medical University, Tianjin, China
| |
Collapse
|
41
|
Chai L, Wu S, Liu G, Wang Z, Tian W, Ma Y. OCILRP2 signaling synergizes with LPS to induce the maturation and differentiation of murine dendritic cells. Biochem Biophys Res Commun 2014; 446:836-42. [PMID: 24631687 DOI: 10.1016/j.bbrc.2014.02.145] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2014] [Accepted: 02/25/2014] [Indexed: 11/29/2022]
Abstract
Osteoclast Inhibitory Lectin-related Protein 2 (OCILRP2) is a typical type II transmembrane protein and belongs to C-type lectin-related protein family. It is preferentially expressed in dendritic cells (DC), B lymphocytes, and activated T lymphocytes. Upon binding to its ligand, OCILRP2 can promote CD28-mediated co-stimulation and enhance T cell activation. However, the role of OCILRP2 in DC development and activation is unclear. In this report, we present evidence that recombinant protein OCILRP2-Fc inhibits the generation and LPS-induced maturation of murine bone marrow-derived dendritic cells (BMDCs) by downregulating the expression of CD11c, MHC-II, and co-stimulators CD80 and CD86. OCILRP2-Fc also reduces the capacity of BMDCs to take up antigens, activates T cells, and secret inflammatory cytokines such as IL-6, IL-12, and TNF-α. Additionally, we show that OCILRP2-Fc may cause the aforementioned effects through inhibiting NF-κB activation. Therefore, OCILRP2 is a new regulator of DC maturation and differentiation following TLR4 activation.
Collapse
Affiliation(s)
- Lihui Chai
- Department of Microbiology and Immunology, Zhengzhou University School of Medicine, 100 Kexue Road, Zhengzhou 450001, People's Republic of China; Laboratory of Cellular and Molecular Immunology, Henan University School of Medicine, The New Campus, Jinming Road, Kaifeng 475004, People's Republic of China
| | - Suxia Wu
- Laboratory of Cellular and Molecular Immunology, Henan University School of Medicine, The New Campus, Jinming Road, Kaifeng 475004, People's Republic of China
| | - Guangchao Liu
- Laboratory of Cellular and Molecular Immunology, Henan University School of Medicine, The New Campus, Jinming Road, Kaifeng 475004, People's Republic of China
| | - Zhanzheng Wang
- Laboratory of Cellular and Molecular Immunology, Henan University School of Medicine, The New Campus, Jinming Road, Kaifeng 475004, People's Republic of China
| | - Wenzhi Tian
- Laboratory of Cellular and Molecular Immunology, Henan University School of Medicine, The New Campus, Jinming Road, Kaifeng 475004, People's Republic of China.
| | - Yuanfang Ma
- Laboratory of Cellular and Molecular Immunology, Henan University School of Medicine, The New Campus, Jinming Road, Kaifeng 475004, People's Republic of China.
| |
Collapse
|
42
|
|
43
|
Role of TREM1-DAP12 in renal inflammation during obstructive nephropathy. PLoS One 2013; 8:e82498. [PMID: 24358193 PMCID: PMC3864959 DOI: 10.1371/journal.pone.0082498] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2013] [Accepted: 10/24/2013] [Indexed: 01/05/2023] Open
Abstract
Tubulo-interstitial damage is a common finding in the chronically diseased kidney and is characterized by ongoing inflammation and fibrosis leading to renal dysfunction and end-stage renal disease. Upon kidney injury, endogenous ligands can be released which are recognized by innate immune sensors to alarm innate immune system. A new family of innate sensors is the family of TREM (triggering receptor expressed on myeloid cell). TREM1 is an activating receptor and requires association with transmembrane adapter molecule DAP12 (DNAX-associated protein 12) for cell signaling. TREM1-DAP12 pathway has a cross-talk with intracellular signaling pathways of several Toll-like receptors (TLRs) and is able to amplify TLR signaling and thereby contributes to the magnitude of inflammation. So far, several studies have shown that TLRs play a role in obstructive nephropathy but the contribution of TREM1-DAP12 herein is unknown. Therefore, we studied TREM1 expression in human and murine progressive renal diseases and further investigated the role for TREM1-DAP12 by subjecting wild-type (WT), TREM1/3 double KO and DAP12 KO mice to murine unilateral ureter obstruction (UUO) model. In patients with hydronephrosis, TREM1 positive cells were observed in renal tissue. We showed that in kidneys from WT mice, DAP12 mRNA and TREM1 mRNA and protein levels were elevated upon UUO. Compared to WT mice, DAP12 KO mice displayed less renal MCP-1, KC and TGF-β1 levels and less influx of macrophages during progression of UUO, whereas TREM1/3 double KO mice displayed less renal MCP-1 level. Renal fibrosis was comparable in WT, TREM1/3 double KO and DAP12 KO mice. We conclude that DAP12, partly through TREM1/3, is involved in renal inflammation during progression of UUO.
Collapse
|
44
|
Xavier-Carvalho C, Gibson G, Brasil P, Ferreira RX, de Souza Santos R, Gonçalves Cruz O, de Oliveira SA, de Sá Carvalho M, Pacheco AG, Kubelka CF, Moraes MO. Single nucleotide polymorphisms in candidate genes and dengue severity in children: A case–control, functional and meta-analysis study. INFECTION GENETICS AND EVOLUTION 2013; 20:197-205. [DOI: 10.1016/j.meegid.2013.08.017] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/08/2013] [Revised: 08/13/2013] [Accepted: 08/21/2013] [Indexed: 12/31/2022]
|
45
|
Thomas CJ, Schroder K. Pattern recognition receptor function in neutrophils. Trends Immunol 2013; 34:317-28. [DOI: 10.1016/j.it.2013.02.008] [Citation(s) in RCA: 129] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2013] [Revised: 02/26/2013] [Accepted: 02/28/2013] [Indexed: 12/13/2022]
|
46
|
Dengue virus therapeutic intervention strategies based on viral, vector and host factors involved in disease pathogenesis. Pharmacol Ther 2013; 137:266-82. [DOI: 10.1016/j.pharmthera.2012.10.007] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2012] [Accepted: 10/15/2012] [Indexed: 12/27/2022]
|
47
|
Abstract
AbstractPersistent high fever is one of the most typical clinical symptoms in dengue virus (DV)–infected patients. However, the source of endogenous pyrogen (eg, IL-1β) and the signaling cascade leading to the activation of inflammasome and caspase-1, which are essential for IL-1β and IL-18 secretion, during dengue infection have not been elucidated yet. Macrophages can be polarized into distinct phenotypes under the influence of GM-CSF or M-CSF, denoted as GM-Mφ and M-Mφ, respectively. We found that DV induced high levels of IL-1β and IL-18 from GM-Mφ (inflammatory macrophage) and caused cell death (pyroptosis), whereas M-Mφ (resting macrophage) did not produce IL-1β and IL-18 on DV infection even with lipopolysaccharide priming. This observation demonstrates the distinct responses of GM-Mφ and M-Mφ to DV infection. Moreover, up-regulation of pro-IL-1β, pro-IL-18, and NLRP3 associated with caspase-1 activation was observed in DV-infected GM-Mφ, whereas blockade of CLEC5A/MDL-1, a C-type lectin critical for dengue hemorrhagic fever and Japanese encephalitis virus infection, inhibits NLRP3 inflammasome activation and pyrotopsis in GM-Mφ. Thus, DV can activate NLRP3 inflammasome via CLEC5A, and GM-Mφ plays a more important role than M-Mφ in the pathogenesis of DV infection.
Collapse
|
48
|
Flipse J, Wilschut J, Smit JM. Molecular mechanisms involved in antibody-dependent enhancement of dengue virus infection in humans. Traffic 2012; 14:25-35. [PMID: 22998156 DOI: 10.1111/tra.12012] [Citation(s) in RCA: 92] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2012] [Revised: 09/19/2012] [Accepted: 09/21/2012] [Indexed: 12/19/2022]
Abstract
Dengue is the most common arthropod-borne viral infection in humans with ∼50 million cases annually worldwide. In recent decades, a steady increase in the number of severe dengue cases has been seen. Severe dengue disease is most often observed in individuals that have pre-existing immunity against heterotypic dengue subtypes and in infants with low levels of maternal dengue antibodies. The generally accepted hypothesis explaining the immunopathogenesis of severe dengue is called antibody-dependent enhancement of dengue infection. Here, circulating antibodies bind to the newly infecting virus but do not neutralize infection. Rather, these antibodies increase the infected cell mass and virus production. Additionally, antiviral responses are diminished allowing massive virus particle production early in infection. The large infected cell mass and the high viral load are prelude for severe disease development. In this review, we discuss what is known about the trafficking of dengue virus in its human host cells, and the signalling pathways activated after virus detection, both in the absence and presence of antibodies against the virus. This review summarizes work that aims to better understand the complex immunopathogenesis of severe dengue disease.
Collapse
Affiliation(s)
- Jacky Flipse
- Department of Medical Microbiology, Molecular Virology Section, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | | | | |
Collapse
|
49
|
Chen ST, Liu RS, Wu MF, Lin YL, Chen SY, Tan DTW, Chou TY, Tsai IS, Li L, Hsieh SL. CLEC5A regulates Japanese encephalitis virus-induced neuroinflammation and lethality. PLoS Pathog 2012; 8:e1002655. [PMID: 22536153 PMCID: PMC3334897 DOI: 10.1371/journal.ppat.1002655] [Citation(s) in RCA: 102] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2011] [Accepted: 03/07/2012] [Indexed: 12/29/2022] Open
Abstract
CLEC5A/MDL-1, a member of the myeloid C-type lectin family expressed on macrophages and neutrophils, is critical for dengue virus (DV)-induced hemorrhagic fever and shock syndrome in Stat1−/− mice and ConA-treated wild type mice. However, whether CLEC5A is involved in the pathogenesis of viral encephalitis has not yet been investigated. To investigate the role of CLEC5A to regulate JEV-induced neuroinflammation, antagonistic anti-CLEC5A mAb and CLEC5A-deficient mice were generated. We find that Japanese encephalitis virus (JEV) directly interacts with CLEC5A and induces DAP12 phosphorylation in macrophages. In addition, JEV activates macrophages to secrete proinflammatory cytokines and chemokines, which are dramatically reduced in JEV-infected Clec5a−/− macrophages. Although blockade of CLEC5A cannot inhibit JEV infection of neurons and astrocytes, anti-CLEC5A mAb inhibits JEV-induced proinflammatory cytokine release from microglia and prevents bystander damage to neuronal cells. Moreover, JEV causes blood-brain barrier (BBB) disintegrity and lethality in STAT1-deficient (Stat1−/−) mice, whereas peripheral administration of anti-CLEC5A mAb reduces infiltration of virus-harboring leukocytes into the central nervous system (CNS), restores BBB integrity, attenuates neuroinflammation, and protects mice from JEV-induced lethality. Moreover, all surviving mice develop protective humoral and cellular immunity against JEV infection. These observations demonstrate the critical role of CLEC5A in the pathogenesis of Japanese encephalitis, and identify CLEC5A as a target for the development of new treatments to reduce virus-induced brain damage. Japanese encephalitis (JE) is one of the most common forms of viral encephalitis worldwide, and the common complication post viral encephalitis is permanent neuropsychiatric sequelae resulting from severe neuroinflammation. However, specific treatment to inhibit JEV-induced neuroinflammation is not available. We found that JEV interacts directly with CLEC5A, a C-type lectin expressed on the myeloid cell surface. This observation led to two major findings; first, we demonstrate that JEV activates macrophages and microglia via CLEC5A, and blockade of CLEC5A reduces bystander neuronal damage and JEV-induced proinflammatory cytokine secretion from macrophages and microglia. Second, peripheral administration of anti-CLEC5A mAb does not only inhibit JEV-induced BBB permeability, but also reduces the numbers of activated microglia and cell infiltration into the CNS. The attenuation of neuronal damage and reduced viral load correlate with the suppression of inflammatory cytokines TNF-α, IL-6, IL-18, and MCP-1 in the CNS. Our studies provide new insights into the molecular mechanism of neuroinflammation, and reveal a possible strategy to control neuroinflammation during viral encephalitis.
Collapse
MESH Headings
- Adaptor Proteins, Signal Transducing/genetics
- Adaptor Proteins, Signal Transducing/immunology
- Adaptor Proteins, Signal Transducing/metabolism
- Animals
- Antibodies, Monoclonal, Murine-Derived/pharmacology
- Astrocytes/immunology
- Astrocytes/metabolism
- Astrocytes/virology
- Blood-Brain Barrier/immunology
- Blood-Brain Barrier/metabolism
- Blood-Brain Barrier/virology
- Cytokines/genetics
- Cytokines/immunology
- Cytokines/metabolism
- Encephalitis Virus, Japanese/genetics
- Encephalitis Virus, Japanese/immunology
- Encephalitis Virus, Japanese/metabolism
- Encephalitis, Japanese/genetics
- Encephalitis, Japanese/immunology
- Encephalitis, Japanese/metabolism
- Immunity, Cellular/genetics
- Immunity, Humoral/genetics
- Inflammation
- Lectins, C-Type/genetics
- Lectins, C-Type/immunology
- Lectins, C-Type/metabolism
- Macrophages/immunology
- Macrophages/metabolism
- Macrophages/virology
- Mice
- Mice, Knockout
- Neurons/immunology
- Neurons/metabolism
- Neurons/virology
- Receptors, Cell Surface/genetics
- Receptors, Cell Surface/immunology
- Receptors, Cell Surface/metabolism
- STAT1 Transcription Factor/genetics
- STAT1 Transcription Factor/immunology
- STAT1 Transcription Factor/metabolism
Collapse
Affiliation(s)
- Szu-Ting Chen
- Department and Institute of Microbiology and Immunology, National Yang-Ming University, Taipei, Taiwan
- Genomics Research Center, Academia Sinica, Taipei, Taiwan
| | - Ren-Shyan Liu
- Molecular and Genetic Imaging Core, Department of Nuclear Medicine, National Yang-Ming University Medical School and Taipei Veterans General Hospital, Taipei, Taiwan
| | - Ming-Fang Wu
- Department and Institute of Microbiology and Immunology, National Yang-Ming University, Taipei, Taiwan
| | - Yi-Ling Lin
- Genomics Research Center, Academia Sinica, Taipei, Taiwan
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Se-Yi Chen
- Department of Neurosurgery Surgical, Taichung Veterans General Hospital, Taichung, Taiwan
- Institute of Clinical Medicine, National Yang-Ming University, Taipei, Taiwan
| | - David Tat-Wei Tan
- Molecular and Genetic Imaging Core, Department of Biomedical Imaging and Radiological Sciences, National Yang-Ming University, Taipei, Taiwan
| | - Teh-Ying Chou
- Institute of Clinical Medicine, National Yang-Ming University, Taipei, Taiwan
| | - I-Shuen Tsai
- Department and Institute of Microbiology and Immunology, National Yang-Ming University, Taipei, Taiwan
| | - Lei Li
- Taipei Blood Center, Taiwan Blood Services Foundation, Taipei, Taiwan
| | - Shie-Liang Hsieh
- Department and Institute of Microbiology and Immunology, National Yang-Ming University, Taipei, Taiwan
- Genomics Research Center, Academia Sinica, Taipei, Taiwan
- Institute of Clinical Medicine, National Yang-Ming University, Taipei, Taiwan
- Infection and Immunity Research Center, National Yang-Ming University, Taipei, Taiwan
- Immunology Center, Taipei Veterans General Hospital, Taipei, Taiwan
- * E-mail:
| |
Collapse
|
50
|
Ward PA. Do MDL-1⁺ cells play a broad role in acute inflammation? J Clin Invest 2011; 121:4234-7. [PMID: 22005298 DOI: 10.1172/jci60122] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Dengue virus (DV) reacts with myeloid DAP12-associating lectin-1 (MDL-1) on immature polymorphonuclear leukocytes. Interaction of DV with MDL-1+ cells triggers systemic inflammatory response syndrome (SIRS) and dengue shock syndrome (DSS), with subsequent multi-organ failure. In this issue of the JCI, Cheung et al. find that sterile acute liver injury in mice is associated with the accumulation of MDL-1+ cells and that triggering of these cells by DV or an MDL-1-specific agonist antibody leads to SIRS, shock, and death. These findings may have broad mechanistic and therapeutic implications for the development of SIRS, sepsis, and shock in humans exposed to a wide array of infectious and non-infectious conditions.
Collapse
Affiliation(s)
- Peter A Ward
- University of Michigan Medical School, Department of Pathology, Ann Arbor, Michigan, USA.
| |
Collapse
|