1
|
Li Q, Huang Y, Ban T, Chen K, Zhen X, Dai Q, Zhang G. Serum amyloid A drive microglia shift to a resolving phenotype through Nrf2. Neuropharmacology 2025; 270:110374. [PMID: 39983914 DOI: 10.1016/j.neuropharm.2025.110374] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Revised: 02/17/2025] [Accepted: 02/18/2025] [Indexed: 02/23/2025]
Abstract
Serum amyloid A (SAA) is an acute-phase protein that has been recognized as a diagnostic biomarker for several diseases. However, the functional studies about the effects of SAA on microglial activation seem controversial. Here, we discovered that SAA induces microglial cells polarize to a pro-resolving M2 phenotype by promoting the stability of the transcription factor Nrf2, which specifically regulates microglia towards a pro-resolving phenotype via metabolic reprogramming. Moreover, we identified that the AMPK/mTOR signaling pathway is involved in SAA-induced Nrf2 upregulation. Additionally, SAA protects cultured neuronal cells from MPP+-induced damage, and furthermore, local administration of SAA into the substantia nigra significantly attenuated MPTP-induced dopaminergic neuronal loss, thereby improving motor impairments in mice. In conclusion, for the first time we demonstrate SAA regulate microglial activation by promoting Nrf2 stabilization, ultimately protecting dopaminergic neurons and alleviating MPTP-induced PD-like pathology.
Collapse
Affiliation(s)
- Qi Li
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and Department of Pharmacology, College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu, 215123, China
| | - Yiwei Huang
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and Department of Pharmacology, College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu, 215123, China
| | - Tao Ban
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and Department of Pharmacology, College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu, 215123, China
| | - Kexin Chen
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and Department of Pharmacology, College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu, 215123, China
| | - Xuechu Zhen
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and Department of Pharmacology, College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu, 215123, China; Join Innovation Center of Hai'an SUDA-, Hai'an Hospital of Traditional Chinese Medicine, Hai'an, Jiangsu, 226600, China
| | - Qijun Dai
- Join Innovation Center of Hai'an SUDA-, Hai'an Hospital of Traditional Chinese Medicine, Hai'an, Jiangsu, 226600, China.
| | - Gufang Zhang
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and Department of Pharmacology, College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu, 215123, China; Join Innovation Center of Hai'an SUDA-, Hai'an Hospital of Traditional Chinese Medicine, Hai'an, Jiangsu, 226600, China.
| |
Collapse
|
2
|
Sanchez NC, Mani G, Nowak JI, Jones C, Kim YI, Sharma NS, Zmijewski JW, Surolia R. Persistent neutrophilic inflammation is associated with delayed toxicity of phenylarsine oxide in lungs. Sci Rep 2025; 15:11840. [PMID: 40195488 PMCID: PMC11976910 DOI: 10.1038/s41598-025-95645-z] [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: 09/17/2024] [Accepted: 03/24/2025] [Indexed: 04/09/2025] Open
Abstract
Phenyl arsine oxide (PAO) is a vesicant, similar to Lewisite, a potential chemical warfare agent and an environmental contaminant. PAO-induced skin burns can trigger acute organ injury, including lungs. We have recently demonstrated that PAO burns can have delayed toxicity, although the specific mechanism/s remain to be determined. A single cutaneous exposure to PAO resulted in inflammatory acute lung injury at 6 and 24 h. While acute injury subsiding by 1 week, we observed a significant airway remodeling at 10 weeks post-PAO exposure. The mechanism of prolonged PAO toxicity was associated with the influx of neutrophils that produced harmful neutrophil extracellular traps (NETs). We demonstrated that the crosstalk between NET deployments and expression of IL-33, a pro-remodeling mediator was associated with the development of peribronchial fibrosis. In summary, these results suggest that a single cutaneous exposure to PAO causes the acute inflammatory phase followed by NETs/IL-33 feed forward signaling implicated for the persistent neutrophil influx and NETs formation resulting in airway remodeling.
Collapse
Affiliation(s)
- Nilda C Sanchez
- Division of Pulmonary, Allergy, and Critical Care, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, 35294, USA
| | - Gopikrishnan Mani
- Division of Pulmonary, Allergy, and Critical Care, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, 35294, USA
| | - Joanna I Nowak
- Division of Pulmonary, Allergy, and Critical Care, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, 35294, USA
| | - Carlin Jones
- Division of Pulmonary, Allergy, and Critical Care, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, 35294, USA
| | - Young-Il Kim
- Division of Pulmonary, Allergy, and Critical Care, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, 35294, USA
| | - Nirmal S Sharma
- Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Jaroslaw W Zmijewski
- Division of Pulmonary, Allergy, and Critical Care, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, 35294, USA
| | - Ranu Surolia
- Division of Pulmonary, Allergy, and Critical Care, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, 35294, USA.
| |
Collapse
|
3
|
Chang Y, Liu Y, Zou Y, Ye RD. Recent Advances in Studies of Serum Amyloid A: Implications in Inflammation, Immunity and Tumor Metastasis. Int J Mol Sci 2025; 26:987. [PMID: 39940756 PMCID: PMC11817213 DOI: 10.3390/ijms26030987] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2024] [Revised: 01/19/2025] [Accepted: 01/22/2025] [Indexed: 02/16/2025] Open
Abstract
Research on serum amyloid A (SAA) has seen major advancement in recent years with combined approaches of structural analysis and genetically altered mice. Initially identified as an acute-phase reactant, SAA is now recognized as a major player in host defense, inflammation, lipid metabolism and tumor metastasis. SAA binding and the neutralization of LPS attenuate sepsis in mouse models. SAA also displays immunomodulatory functions in Th17 differentiation and macrophage polarization, contributing to a pro-metastatic tumor microenvironment. In spite of the progress, the regulatory mechanisms for these diverse functions of SAA remain unclear. This review provides a brief summary of recent advances in SAA research on immunity, inflammation, tumor microenvironment and in vivo models.
Collapse
Affiliation(s)
- Yixin Chang
- Kobilka Institute of Innovative Drug Discovery, School of Medicine, The Chinese University of Hong Kong, Shenzhen 518172, China
| | - Yezhou Liu
- Kobilka Institute of Innovative Drug Discovery, School of Medicine, The Chinese University of Hong Kong, Shenzhen 518172, China
| | - Yuanrui Zou
- Kobilka Institute of Innovative Drug Discovery, School of Medicine, The Chinese University of Hong Kong, Shenzhen 518172, China
| | - Richard D. Ye
- Kobilka Institute of Innovative Drug Discovery, School of Medicine, The Chinese University of Hong Kong, Shenzhen 518172, China
- The Chinese University of Hong Kong, Shenzhen Futian Biomedical Innovation R&D Center, Shenzhen 518000, China
| |
Collapse
|
4
|
Sanchez NC, Mani G, Jones C, Zmijewsli JW, Surolia R. Persistent Neutrophilic Inflammation is Associated with Delayed Toxicity of Phenylarsine Oxide in Lungs. RESEARCH SQUARE 2025:rs.3.rs-5100050. [PMID: 39877099 PMCID: PMC11774458 DOI: 10.21203/rs.3.rs-5100050/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/31/2025]
Abstract
Phenyl arsine oxide (PAO) is a vesicant, similar to Lewisite, a potential chemical warfare agent and an environmental contaminant. PAO-induced skin burns can trigger acute organ injury, including lungs. We have recently demonstrated that PAO burns can also has a delayed toxicity, although the specific mechanism/s remain to be determined. A single cutaneous exposure to PAO resulted in inflammatory acute lung injury at 6 and 24 hours. While acute injury subsiding by 1 week, we observed a significant airway remodeling at 10 weeks post-PAO exposure. The mechanism of prolonged PAO toxicity was associated with the influx of neutrophils that produced harmful neutrophil extracellular traps (NETs). We demonstrated that the crosstalk between NET deployments and expression of IL-33, a pro-remodeling mediator was associated with the development of peribronchial fibrosis. In summary, these results suggest that a single cutaneous exposure to PAO causes the acute inflammatory phase followed by NETs/IL-33 feed forward signaling implicated for the persistent neutrophil influx and NETs formation resulting in airway remodeling.
Collapse
|
5
|
Ahmad P, Hussain A, Siqueira WL. Mass spectrometry-based proteomic approaches for salivary protein biomarkers discovery and dental caries diagnosis: A critical review. MASS SPECTROMETRY REVIEWS 2024; 43:826-856. [PMID: 36444686 DOI: 10.1002/mas.21822] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
Dental caries is a multifactorial chronic disease resulting from the intricate interplay among acid-generating bacteria, fermentable carbohydrates, and several host factors such as saliva. Saliva comprises several proteins which could be utilized as biomarkers for caries prevention, diagnosis, and prognosis. Mass spectrometry-based salivary proteomics approaches, owing to their sensitivity, provide the opportunity to investigate and unveil crucial cariogenic pathogen activity and host indicators and may demonstrate clinically relevant biomarkers to improve caries diagnosis and management. The present review outlines the published literature of human clinical proteomics investigations on caries and extensively elucidates frequently reported salivary proteins as biomarkers. This review also discusses important aspects while designing an experimental proteomics workflow. The protein-protein interactions and the clinical relevance of salivary proteins as biomarkers for caries, together with uninvestigated domains of the discipline are also discussed critically.
Collapse
Affiliation(s)
- Paras Ahmad
- College of Dentistry, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | - Ahmed Hussain
- College of Dentistry, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | - Walter L Siqueira
- College of Dentistry, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| |
Collapse
|
6
|
He PY, Wu MY, Zheng LY, Duan Y, Fan Q, Zhu XM, Yao YM. Interleukin-33/serum stimulation-2 pathway: Regulatory mechanisms and emerging implications in immune and inflammatory diseases. Cytokine Growth Factor Rev 2024; 76:112-126. [PMID: 38155038 DOI: 10.1016/j.cytogfr.2023.12.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Accepted: 12/11/2023] [Indexed: 12/30/2023]
Abstract
Interleukin (IL)- 33, a nuclear factor and pleiotropic cytokine of the IL-1 family, is gaining attention owing to its important role in chronic inflammatory and autoimmune diseases. This review extends our knowledge of the effects exerted by IL-33 on target cells by binding to its specific receptor serum stimulation-2 (ST2). Depending on the tissue context, IL-33 performs multiple functions encompassing host defence, immune response, initiation and amplification of inflammation, tissue repair, and homeostasis. The levels and activity of IL-33 in the body are controlled by complex IL-33-targeting regulatory pathways. The unique temporal and spatial expression patterns of IL-33 are associated with host homeostasis and the development of immune and inflammatory disorders. Therefore, understanding the origin, function, and processes of IL-33 under various conditions is crucial. This review summarises the regulatory mechanisms underlying the IL-33/ST2 signalling axis and its potential role and clinical significance in immune and inflammatory diseases, and discusses the current complex and conflicting findings related to IL-33 in host responses.
Collapse
Affiliation(s)
- Peng-Yi He
- Translational Medicine Research Center, Medical Innovation Research Division and Fourth Medical Center of the Chinese PLA General Hospital, Beijing 100853, China; School of Medicine, Nankai University, Tianjin 300071, China
| | - Meng-Yao Wu
- Translational Medicine Research Center, Medical Innovation Research Division and Fourth Medical Center of the Chinese PLA General Hospital, Beijing 100853, China
| | - Li-Yu Zheng
- Translational Medicine Research Center, Medical Innovation Research Division and Fourth Medical Center of the Chinese PLA General Hospital, Beijing 100853, China
| | - Yu Duan
- Translational Medicine Research Center, Medical Innovation Research Division and Fourth Medical Center of the Chinese PLA General Hospital, Beijing 100853, China
| | - Qi Fan
- Emergency Medicine Center, First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325015, China
| | - Xiao-Mei Zhu
- Tissue Repair and Regeneration Research Center, Medical Innovation Research Division and Fourth Medical Center of the Chinese PLA General Hospital, Beijing 100048, China.
| | - Yong-Ming Yao
- Translational Medicine Research Center, Medical Innovation Research Division and Fourth Medical Center of the Chinese PLA General Hospital, Beijing 100853, China; School of Medicine, Nankai University, Tianjin 300071, China.
| |
Collapse
|
7
|
Yang J, Yang K, Wang K, Zhou D, Zhou J, Du X, Liu S, Cheng Z. Serum amyloid A regulates TLR2/4-mediated IFN-β signaling pathway against Marek's disease virus. Virus Res 2023; 326:199044. [PMID: 36652973 DOI: 10.1016/j.virusres.2023.199044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Revised: 01/09/2023] [Accepted: 01/13/2023] [Indexed: 01/16/2023]
Abstract
Serum amyloid A (SAA), an acute response phase protein (APP), is crucial for the innate immune response during pathogenic microorganisms' invasion. Marek's disease virus (MDV) is a highly oncogenic alphaherpesvirus that activates multiple innate immune molecules, including SAA, in the host during infection. However, the pathway through which SAA participates in MDV-induced host innate immunity remains unknown. The present study aimed to elucidate the pathway through which SAA exerts its anti-MDV function. We observed that MDV infection in vivo and in vitro significantly elevated SAA expression. Furthermore, through SAA overexpression and knockdown experiments, we demonstrated that SAA could inhibit MDV replication. Subsequently, we found that SAA activated Toll-Like Receptor 2/4 (TLR2/4) -mediated Interferon Beta (IFN-β) promoter activity and IFN regulatory factor 7 (IRF7) promoter activity. During MDV infection, SAA enhanced TLR2/4-mediated IFN-β signal transduction and messenger RNAs (mRNAs) expression of type I IFN (IFN-I) and interferon-stimulated genes (ISGs). Finally, TLR2/4 inhibitor OxPAPC inhibits the anti-MDV activity of SAA. These results demonstrated that SAA inhibits MDV replication and enhancing TLR2/4-mediated IFN-β signal transduction to promote IFNs and ISGs expression. This finding is the first to demonstrate the signaling pathway by which SAA exerts its anti-MDV function. It also provides new insights into the control of oncogenic herpesviruses from the perspective of acute response phase proteins.
Collapse
Affiliation(s)
- Jianhao Yang
- College of Veterinary Medicine, Shandong Agricultural University, 61 Daizong Street, Taian 271018, China
| | - Kunmei Yang
- College of Veterinary Medicine, Shandong Agricultural University, 61 Daizong Street, Taian 271018, China
| | - Kang Wang
- College of Veterinary Medicine, Shandong Agricultural University, 61 Daizong Street, Taian 271018, China
| | - Defang Zhou
- College of Veterinary Medicine, Shandong Agricultural University, 61 Daizong Street, Taian 271018, China
| | - Jing Zhou
- College of Veterinary Medicine, Shandong Agricultural University, 61 Daizong Street, Taian 271018, China
| | - Xusheng Du
- College of Veterinary Medicine, Shandong Agricultural University, 61 Daizong Street, Taian 271018, China
| | - Shenglong Liu
- College of Veterinary Medicine, Shandong Agricultural University, 61 Daizong Street, Taian 271018, China
| | - Ziqiang Cheng
- College of Veterinary Medicine, Shandong Agricultural University, 61 Daizong Street, Taian 271018, China.
| |
Collapse
|
8
|
Demyanets S, Stojkovic S, Huber K, Wojta J. The Paradigm Change of IL-33 in Vascular Biology. Int J Mol Sci 2021; 22:ijms222413288. [PMID: 34948083 PMCID: PMC8707059 DOI: 10.3390/ijms222413288] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Revised: 11/30/2021] [Accepted: 12/07/2021] [Indexed: 12/30/2022] Open
Abstract
In this review, we focus on the actual understanding of the role of IL-33 in vascular biology in the context of the historical development since the description of IL-33 as a member of IL-1 superfamily and the ligand for ST2 receptor in 2005. We summarize recent data on the biology, structure and signaling of this dual-function factor with both nuclear and extracellular cytokine properties. We describe cellular sources of IL-33, particularly within vascular wall, changes in its expression in different cardio-vascular conditions and mechanisms of IL-33 release. Additionally, we summarize the regulators of IL-33 expression as well as the effects of IL-33 itself in cells of the vasculature and in monocytes/macrophages in vitro combined with the consequences of IL-33 modulation in models of vascular diseases in vivo. Described in murine atherosclerosis models as well as in macrophages as an atheroprotective cytokine, extracellular IL-33 induces proinflammatory, prothrombotic and proangiogenic activation of human endothelial cells, which are processes known to be involved in the development and progression of atherosclerosis. We, therefore, discuss that IL-33 can possess both protective and harmful effects in experimental models of vascular pathologies depending on experimental conditions, type and dose of administration or method of modulation.
Collapse
Affiliation(s)
- Svitlana Demyanets
- Department of Laboratory Medicine, Medical University of Vienna, 1090 Vienna, Austria;
| | - Stefan Stojkovic
- Department of Internal Medicine II, Division of Cardiology, Medical University of Vienna, 1090 Vienna, Austria;
| | - Kurt Huber
- 3rd Medical Department with Cardiology and Intensive Care Medicine, Clinic Ottakring, 1160 Vienna, Austria;
- Medical School, Sigmund Freud University, 1020 Vienna, Austria
- Ludwig Boltzmann Institute for Cardiovascular Research, 1090 Vienna, Austria
| | - Johann Wojta
- Department of Internal Medicine II, Division of Cardiology, Medical University of Vienna, 1090 Vienna, Austria;
- Ludwig Boltzmann Institute for Cardiovascular Research, 1090 Vienna, Austria
- Core Facilities, Medical University of Vienna, 1090 Vienna, Austria
- Correspondence: ; Tel.: +43-1-40400-73500; Fax: +43-1-40400-73586
| |
Collapse
|
9
|
Zheng J, Yao L, Zhou Y, Gu X, Wang C, Bao K, Sun Y, Hong M. A novel function of NLRP3 independent of inflammasome as a key transcription factor of IL-33 in epithelial cells of atopic dermatitis. Cell Death Dis 2021; 12:871. [PMID: 34561424 PMCID: PMC8463682 DOI: 10.1038/s41419-021-04159-9] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Revised: 08/24/2021] [Accepted: 09/03/2021] [Indexed: 12/25/2022]
Abstract
Atopic dermatitis (AD) is a common chronic pruritic inflammatory skin disorder characterized by recurrent eczematous lesions. Interleukin (IL)-33, a cytokine of the IL-1 family, was found to play an important role in the pathogenesis of AD. As a key component of the inflammasome, NLRP3 has been mostly described in myeloid cells that to mediate inflammasome activation conducted proinflammatory cytokine production of the IL-1 family. However, the role of NLRP3 inflammasome in the pathogenesis of AD, as well as IL-33 processing are highly controversial. Whether NLRP3 can mediate IL-33 expression and secretion independently of the inflammasome in the epithelium of AD has remained unclear. In this article, we found the mRNA expression of Il33 and Nlrp3 were notably increased in the lesional skin of AD patients compared to healthy controls. We then found a significant positive correlation between the expression of Nlrp3 and Il33 in the epithelium of MC903-mediated AD mice model, but no changes were observed for Il36α, Il36γ, Il1β, or Il18 mRNA expression, as well as IL-1β or IL-18 production. Overexpression of NLRP3 in human immortalized epithelial cells increased IL-33 expression, whereas siRNA targeting NLRP3 abolished IL-33 expression. In addition, inhibition of NLRP3 inflammasome activation or caspase-1 activity with MCC950 or VX-765 showed no effect on the expression and secretion of IL-33 in AD mice. Unlike myeloid cells, NLRP3 predominantly located in the nucleus of epithelial cells, which could directly bind to Il33 specific-promoters and transactivate it through an interaction with transcription factor IRF4. Furthermore, NLRP3 deficient mice exhibited a significant alleviated epidermis inflammation and decreased mRNA expression and secretion of IL-33 in MC903-mediated AD mice without interfering with TSLP and IL-1β production. Our results demonstrate a novel ability of NLRP3 to function as a crucial transcription factor of IL-33 in epithelium independently of inflammasome that to mediate the pathological process of AD.
Collapse
MESH Headings
- Animals
- Calcitriol/analogs & derivatives
- Cell Nucleus/metabolism
- Dermatitis, Atopic/genetics
- Dermatitis, Atopic/metabolism
- Dermatitis, Atopic/pathology
- Disease Models, Animal
- Epithelial Cells/metabolism
- Gene Expression Regulation
- HaCaT Cells
- Humans
- Inflammasomes/metabolism
- Interferon Regulatory Factors/metabolism
- Interleukin-33/metabolism
- Male
- Mice, Inbred BALB C
- Mice, Inbred C57BL
- NLR Family, Pyrin Domain-Containing 3 Protein/deficiency
- NLR Family, Pyrin Domain-Containing 3 Protein/metabolism
- Protein Binding
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- Transcription Factors/metabolism
- Mice
Collapse
Affiliation(s)
- Jie Zheng
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, 138 Xianlin Avenue, Nanjing, 210023, China
- Department of Pharmacology, School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, 138 Xianlin Avenue, Nanjing, 210023, China
| | - Lu Yao
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, 138 Xianlin Avenue, Nanjing, 210023, China
- Department of Biotherapy, Nanjing Jinling Hospital, Nanjing, 210002, China
| | - Yijing Zhou
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, 138 Xianlin Avenue, Nanjing, 210023, China
| | - Xiaoqun Gu
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, 138 Xianlin Avenue, Nanjing, 210023, China
| | - Can Wang
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, 138 Xianlin Avenue, Nanjing, 210023, China
| | - Kaifan Bao
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, 138 Xianlin Avenue, Nanjing, 210023, China
- Department of Immunology, School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, 138 Xianlin Avenue, Nanjing, 210023, China
| | - Yang Sun
- State Key Laboratory of Pharmaceutical Biotechnology, Chemistry and Biomedicine Innovation Center, School of Life Sciences, Nanjing University, 163 Xianlin Avenue, Nanjing, 210023, China
| | - Min Hong
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, 138 Xianlin Avenue, Nanjing, 210023, China.
| |
Collapse
|
10
|
Xu S, Mei S, Lu J, Wu H, Dong X, Shi L, Zhou J, Zhang J. Transcriptome Analysis of Microglia Reveals That the TLR2/IRF7 Signaling Axis Mediates Neuroinflammation After Subarachnoid Hemorrhage. Front Aging Neurosci 2021; 13:645649. [PMID: 34276335 PMCID: PMC8278202 DOI: 10.3389/fnagi.2021.645649] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Accepted: 04/19/2021] [Indexed: 12/23/2022] Open
Abstract
Microglia-mediated neuroinflammatory response in the early brain injury after subarachnoid hemorrhage (SAH) has been reported to have an impact on progress, and the mechanism is not completely understood. Here, we performed genome-wide transcriptome analysis of microglia purified from damaged hemisphere of adult mice at 3 days after SAH or sham operation. Robust transcriptional changes were observed between SAH-induced and healthy microglia, indicating rapid activation of microglia after suffering from SAH. We identified 1576 differentially expressed genes (DEGs; 928 upregulated and 648 downregulated) in SAH-induced microglia compared with sham microglia, representing a strong alteration of the genome (6.85% of total ∼23,000 genes). Functional enrichment of these DEGs indicated that cell division, inflammatory response, cytokine production, and leukocyte chemotaxis were strongly activated in SAH-induced microglia. Moreover, we identified and proved that the TLR2/IRF7 signaling axis was involved in the regulation of this microglia-mediated inflammation in SAH mice by performing flow cytometry and immunofluorescence. Together, these results provided a perspective of microglia-mediated neuroinflammatory response in the early stage of SAH and might give a new therapeutic target for SAH.
Collapse
Affiliation(s)
- Shenbin Xu
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Shuhao Mei
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Jianan Lu
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Haijian Wu
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Xiao Dong
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Ligen Shi
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Jingyi Zhou
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Jianmin Zhang
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China.,Brain Research Institute, Zhejiang University, Hangzhou, China.,Collaborative Innovation Center for Brain Science, Zhejiang University, Hangzhou, China
| |
Collapse
|
11
|
Influence of obesity on serum levels of SARS-CoV-2-specific antibodies in COVID-19 patients. PLoS One 2021; 16:e0245424. [PMID: 33760825 PMCID: PMC7990309 DOI: 10.1371/journal.pone.0245424] [Citation(s) in RCA: 54] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Accepted: 02/25/2021] [Indexed: 12/13/2022] Open
Abstract
SARS-CoV-2 (Severe Acute Respiratory Syndrome Corona Virus-2), cause of COVID-19 (Coronavirus Disease of 2019), represents a significant risk to people living with pre-existing conditions associated with exacerbated inflammatory responses and consequent dysfunctional immunity. In this paper, we have evaluated the influence of obesity, a condition associated with chronic systemic inflammation, on the secretion of SARS-CoV-2-specific IgG antibodies in the blood of COVID-19 patients. Our hypothesis is that obesity is associated with reduced amounts of specific IgG antibodies. Results have confirmed our hypothesis and have shown that SARS-CoV-2 IgG antibodies are negatively associated with Body Mass Index (BMI) in COVID-19 obese patients, as expected based on the known influence of obesity on humoral immunity. Antibodies in COVID-19 obese patients are also negatively associated with serum levels of pro-inflammatory and metabolic markers of inflammaging and pulmonary inflammation, such as SAA (serum amyloid A protein), CRP (C-reactive protein), and ferritin, but positively associated with NEFA (nonesterified fatty acids). These results altogether could help to identify an inflammatory signature with strong predictive value for immune dysfunction. Inflammatory markers identified may subsequently be targeted to improve humoral immunity in individuals with obesity and in individuals with other chronic inflammatory conditions.
Collapse
|
12
|
Frasca D, Reidy L, Cray C, Diaz A, Romero M, Kahl K, Blomberg BB. Effects of obesity on serum levels of SARS-CoV-2-specific antibodies in COVID-19 patients. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2020:2020.12.18.20248483. [PMID: 33403370 PMCID: PMC7783955 DOI: 10.1101/2020.12.18.20248483] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/13/2023]
Abstract
SARS-CoV-2 (Severe Acute Respiratory Syndrome Corona Virus-2), cause of COVID-19 (Coronavirus Disease of 2019), represents a significant risk to people living with pre-existing conditions associated with exacerbated inflammatory responses and consequent dysfunctional immunity. In this paper, we have evaluated the effects of obesity, a condition associated with chronic systemic inflammation, on the secretion of SARS-CoV-2-specific IgG antibodies in the blood of COVID-19 patients. Results have shown that SARS-CoV-2 IgG antibodies are negatively associated with Body Mass Index (BMI) in COVID-19 obese patients, as expected based on the known effects of obesity on humoral immunity. Antibodies in COVID-19 obese patients are also negatively associated with serum levels of pro-inflammatory and metabolic markers of inflammaging and pulmonary inflammation, such as SAA (serum amyloid A protein), CRP (C-reactive protein) and ferritin, but positively associated with NEFA (nonesterified fatty acids). These results altogether could help to identify an inflammatory signature with strong predictive value for immune dysfunction that could be targeted to improve humoral immunity in individuals with obesity as well as with other chronic inflammatory conditions.
Collapse
|
13
|
White MR, Hsieh IN, De Luna X, Hartshorn KL. Effects of serum amyloid protein A on influenza A virus replication and viral interactions with neutrophils. J Leukoc Biol 2020; 110:155-166. [PMID: 33205458 PMCID: PMC7753654 DOI: 10.1002/jlb.4ab0220-116rr] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Revised: 10/13/2020] [Accepted: 10/26/2020] [Indexed: 01/22/2023] Open
Abstract
Innate immunity is vital for the early control of influenza A virus (IAV) infection. Serum amyloid A (SAA1) is an acute phase reactant produced in the liver and lung that rises dramatically during IAV infection. The potential role of SAA1 in host defense against IAV is unknown. SAA1 has been reported to directly activate neutrophils and to recruit them to the lung during infectious and inflammatory processes. Neutrophils are the most abundant cell recruited to the lung in the early phase of IAV infection. There are different forms and preparations of SAA1 that have found to have different effects on phagocyte responses, through various receptors. In this paper, we test the direct effects of various preparations of serum derived or recombinant SAA on IAV and how it modulates the interactions of IAV with neutrophils. All SAA preparations bound to IAV in vitro but caused minimal hemagglutination inhibition or viral aggregation. The human serum‐derived SAA1 or the complex of SAA1 with HDL did have IAV neutralizing activity in vitro, whereas the recombinant SAA1 preparations did not. We found that different SAA preparations also had markedly different effects on neutrophil functions, with E. coli‐derived SAA1 triggering some responses in neutrophils on its own or in presence of IAV whereas mammalian cell‐derived SAA1 did not. This discrepancy could be explained by the reported contamination of the former preparation with bacterial components. Of interest, however, serum SAA alone, serum SAA complexed with HDL, or HDL alone potentiated some neutrophil responses to IAV. Our results suggest that SAA may play some role in host response to IAV, but further work needs to be done to clarify the role of different variants of SAA alone or complexed with HDL.
Collapse
Affiliation(s)
- Mitchell R White
- Department of Medicine, Boston University School of Medicine, Boston, Massachusetts, USA
| | - I-Ni Hsieh
- Department of Medicine, Boston University School of Medicine, Boston, Massachusetts, USA
| | - Xavier De Luna
- Department of Medicine, Boston University School of Medicine, Boston, Massachusetts, USA
| | - Kevan L Hartshorn
- Department of Medicine, Boston University School of Medicine, Boston, Massachusetts, USA
| |
Collapse
|
14
|
Yang X, Li R, Xu L, Qian F, Sun L. Serum amyloid A3 is required for caerulein-induced acute pancreatitis through induction of RIP3-dependent necroptosis. Immunol Cell Biol 2020; 99:34-48. [PMID: 32725692 DOI: 10.1111/imcb.12382] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2019] [Revised: 03/28/2020] [Accepted: 07/23/2020] [Indexed: 02/06/2023]
Abstract
Serum amyloid A (SAA) is an early and sensitive biomarker of inflammatory diseases, but its role in acute pancreatitis (AP) is still unclear. Here, we used a caerulein-induced mouse model to investigate the role of SAA in AP and other related inflammatory responses. In our study, we found that the expression of a specific SAA isoform, SAA3, was significantly elevated in a caerulein-induced AP animal model. In addition, SAA3-knockout (Saa3-/- ) mice showed lower serum levels of amylase and lipase, tissue damage and proinflammatory cytokine production in the pancreas compared with those of wild-type mice in response to caerulein administration. AP-associated acute lung injury was also significantly attenuated in Saa3-/- mice. In our in vitro experiments, treatment with cholecystokinin and recombinant SAA3 significantly induced necroptosis and cytokine production. Moreover, we found that the regulatory effect of SAA3 on acinar cell necroptosis was through a receptor-interacting protein 3 (RIP3)-dependent manner. Collectively, our findings indicate that SAA3 is required for AP by inducing an RIP3-dependent necroptosis pathway in acinar cells and is a potential drug target for AP.
Collapse
Affiliation(s)
- Xinyi Yang
- Engineering Research Center of Cell & Therapeutic Antibody, Ministry of Education, School of Pharmacy, Shanghai Jiao Tong University, Shanghai, 200240, PR China
| | - Runsheng Li
- Department of Hematology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, 301 Yanchang Road, Shanghai, 200072, PR China
| | - Lu Xu
- Engineering Research Center of Cell & Therapeutic Antibody, Ministry of Education, School of Pharmacy, Shanghai Jiao Tong University, Shanghai, 200240, PR China
| | - Feng Qian
- Engineering Research Center of Cell & Therapeutic Antibody, Ministry of Education, School of Pharmacy, Shanghai Jiao Tong University, Shanghai, 200240, PR China.,Anhui Province Key Laboratory of Translational Cancer Research, Bengbu Medical College, Anhui Province, Bengbu, 233003, PR China
| | - Lei Sun
- Engineering Research Center of Cell & Therapeutic Antibody, Ministry of Education, School of Pharmacy, Shanghai Jiao Tong University, Shanghai, 200240, PR China
| |
Collapse
|
15
|
Guedes SFF, Neves BG, Bezerra DS, Souza GHMF, Lima-Neto ABM, Guedes MIF, Duarte S, Rodrigues LKA. Saliva proteomics from children with caries at different severity stages. Oral Dis 2020; 26:1219-1229. [PMID: 32285988 DOI: 10.1111/odi.13352] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2019] [Revised: 03/18/2020] [Accepted: 03/27/2020] [Indexed: 12/11/2022]
Abstract
OBJECTIVE To perform a comparative analysis of saliva protein profile of patients with early childhood caries at different levels of severity and caries-free individuals. MATERIALS AND METHODS Stimulated saliva samples were collected from 126 children (2-6 years old), classified according to the ICDAS II, and divided into 3 groups (n = 42): caries-free (CF), enamel caries (EC), and dentine caries (DC). Samples were digested and analyzed by nanoUPLC coupled with a mass spectrometry. Data analyses were conducted with Progenesis QI for Proteomics Software v2.0. Gene Ontology (GO) terms and protein-protein interaction analysis were obtained. RESULTS A total of 306 proteins (≈6 peptides) were identified. Among them, 122 were differentially expressed in comparisons among children with different caries status. Out of the 122 proteins, the proteins E2AK4 and SH3L2 were exclusively present in groups CF and EC, respectively, and 8 proteins (HAUS4, CAH1, IL36A, IL36G, AIMP1, KLHL8, KLH13, and SAA1) were considered caries-related proteins when compared to caries-free children; they were up-regulated proteins in the caries groups (EC and DC). CONCLUSION The identification of exclusive proteins for caries-free or carious-related conditions may help in understanding the mechanisms of caries and predicting risk as well as advancing in caries control or anti-caries approaches.
Collapse
Affiliation(s)
- Sarah F F Guedes
- Faculty of Pharmacy, Dentistry and Nursing, Postgraduate Program in Dentistry, Federal University of Ceará, Fortaleza, Brazil
| | - Beatriz G Neves
- School of Dentistry, Federal University of Ceará, Sobral, Brazil
| | | | - Gustavo H M F Souza
- MS Applications Development Laboratory, Waters Corporation, São Paulo, Brazil
| | - Abelardo B M Lima-Neto
- Laboratory of Biotechnology and Molecular Biology, State University of Ceará, Fortaleza, Brazil
| | - Maria Izabel F Guedes
- Laboratory of Biotechnology and Molecular Biology, State University of Ceará, Fortaleza, Brazil
| | - Simone Duarte
- Department of Cariology, Operative Dentistry and Dental Public Health, School of Dentistry, Indiana University - Purdue University Indianapolis, Indianapolis, IN, USA
| | - Lidiany K A Rodrigues
- Faculty of Pharmacy, Dentistry and Nursing, Postgraduate Program in Dentistry, Federal University of Ceará, Fortaleza, Brazil
| |
Collapse
|
16
|
Minaga K, Watanabe T, Arai Y, Shiokawa M, Hara A, Yoshikawa T, Kamata K, Yamashita K, Kudo M. Activation of interferon regulatory factor 7 in plasmacytoid dendritic cells promotes experimental autoimmune pancreatitis. J Gastroenterol 2020; 55:565-576. [PMID: 31960143 DOI: 10.1007/s00535-020-01662-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/06/2019] [Accepted: 01/05/2020] [Indexed: 02/04/2023]
Abstract
BACKGROUND Excessive type I IFN (IFN-I) production by plasmacytoid dendritic cells (pDCs) promotes autoimmunity. Recently, we reported that a prominent feature of both experimental autoimmune pancreatitis (AIP) and human type 1 AIP is pDC activation followed by enhanced production of IFN-I and IL-33. However, the roles played by interferon regulatory factor 7 (IRF7), a critical transcription factor for IFN-I production in pDCs, in these disorders have not been clarified. METHODS Whole and nuclear extracts were isolated from pancreatic mononuclear cells (PMNCs) from MRL/MpJ mice exhibiting AIP. Expression of phospho-IRF7 and nuclear translocation of IRF7 was examined in these extracts by immunoblotting. Pancreatic expression of IRF7 was assessed by immunofluorescence analysis in experimental AIP. Nuclear translocation of IRF7 upon exposure to neutrophil extracellular traps (NETs) was assessed in peripheral blood pDCs from type 1 AIP patients. Pancreatic IRF7 expression was examined in surgically operated specimens from type 1 AIP patients. RESULTS IRF7 activation was induced in pancreatic pDCs in experimental AIP. siRNA-mediated knockdown of IRF7 expression prevented AIP development, which was accompanied by a marked reduction in both pancreatic accumulation of pDCs and production of IFN-α and IL-33. Notably, in peripheral blood pDCs isolated from patients with type 1 AIP, nuclear translocation of IRF7 was enhanced as compared with the translocation in pDCs from healthy controls. Furthermore, IRF7-expressing pDCs were detected in the pancreas of patients with type 1 AIP. CONCLUSIONS These findings suggest that the IRF7-IFN-I-IL-33 axis activated in pDCs drives pathogenic innate immune responses associated with type 1 AIP.
Collapse
Affiliation(s)
- Kosuke Minaga
- Department of Gastroenterology and Hepatology, Kindai University Faculty of Medicine, 377-2 Ohno-Higashi, Osaka-Sayama, Osaka, 589-8511, Japan
| | - Tomohiro Watanabe
- Department of Gastroenterology and Hepatology, Kindai University Faculty of Medicine, 377-2 Ohno-Higashi, Osaka-Sayama, Osaka, 589-8511, Japan.
| | - Yasuyuki Arai
- Department of Hematology and Oncology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Masahiro Shiokawa
- Department of Gastroenterology and Hepatology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Akane Hara
- Department of Gastroenterology and Hepatology, Kindai University Faculty of Medicine, 377-2 Ohno-Higashi, Osaka-Sayama, Osaka, 589-8511, Japan
| | - Tomoe Yoshikawa
- Department of Gastroenterology and Hepatology, Kindai University Faculty of Medicine, 377-2 Ohno-Higashi, Osaka-Sayama, Osaka, 589-8511, Japan
| | - Ken Kamata
- Department of Gastroenterology and Hepatology, Kindai University Faculty of Medicine, 377-2 Ohno-Higashi, Osaka-Sayama, Osaka, 589-8511, Japan
| | - Kouhei Yamashita
- Department of Hematology and Oncology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Masatoshi Kudo
- Department of Gastroenterology and Hepatology, Kindai University Faculty of Medicine, 377-2 Ohno-Higashi, Osaka-Sayama, Osaka, 589-8511, Japan
| |
Collapse
|
17
|
Picroside II Isolated from Pseudolysimachion rotundum var. subintegrum Inhibits Glucocorticoid Refractory Serum Amyloid A (SAA) Expression and SAA-induced IL-33 Secretion. Molecules 2019; 24:molecules24102020. [PMID: 31137813 PMCID: PMC6572537 DOI: 10.3390/molecules24102020] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2019] [Revised: 05/23/2019] [Accepted: 05/23/2019] [Indexed: 11/29/2022] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is a major inflammatory lung disease characterized by irreversible and progressive airflow obstruction. Although corticosteroids are often used to reduce inflammation, steroid therapies are insufficient in patients with refractory COPD. Both serum amyloid A (SAA) and IL-33 have been implicated in the pathology of steroid-resistant lung inflammation. Picroside II isolated from Pseudolysimachion rotundum var. subintegrum(Plantaginaceae) is a major bioactive component of YPL-001, which has completed phase-2a clinical trials in chronic obstructive pulmonary disease patients. In this study, we investigated whether picroside II is effective in treating steroid refractory lung inflammation via the inhibition of the SAA-IL-33 axis. Picroside II inhibited LPS-induced SAA1 expression in human monocytes, which are resistant to steroids. SAA induced the secretion of IL-33 without involving cell necrosis. Picroside II, but not dexamethasone effectively inhibited SAA-induced IL-33 expression and secretion. The inhibitory effect by picroside II was mediated by suppressing the mitogen-activated protein kinase (MAPK) p38, ERK1/2, and nuclear factor-κB pathways. Our results suggest that picroside II negatively modulates the SAA-IL-33 axis that has been implicated in steroid-resistant lung inflammation. These findings provide valuable information for the development of picroside II as an alternative therapeutic agent against steroid refractory lung inflammation in COPD.
Collapse
|
18
|
Wang YW, Wang WS, Wang LY, Bao YR, Lu JW, Lu Y, Zhang CY, Li WJ, Sun K, Ying H. Extracellular matrix remodeling effects of serum amyloid A1 in the human amnion: Implications for fetal membrane rupture. Am J Reprod Immunol 2019; 81:e13073. [PMID: 30461130 DOI: 10.1111/aji.13073] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2018] [Revised: 10/30/2018] [Accepted: 11/15/2018] [Indexed: 12/14/2022] Open
Abstract
PROBLEM Rupture of fetal membranes is a crucial event at parturition, which is preceded by extensive extracellular matrix (ECM) remodeling. Our recent studies have demonstrated that the human fetal membranes are capable of de novo synthesis of serum amyloid A1 (SAA1), an acute phase protein, and the abundance of SAA1 in the amnion was increased at parturition. However, the exact role of SAA1 in human parturition remains to be established. METHOD OF STUDY The effects of SAA1 on the abundance of collagenases and lysyl oxidase, the enzyme that cross-links collagens, were investigated in culture primary human amnion fibroblasts and tissue explants with an aim to examine the involvement of SAA1 in the ECM remodeling in the amnion. RESULTS Serum amyloid A1 (SAA1) time- and dose-dependently increased the abundance of collagenases MMP-1, MMP-8, and MMP-13, while decreased the abundance of lysyl oxidase-like 1 (LOXL1). These effects of SAA1 were attenuated by siRNA-mediated knockdown of the Toll-like receptor (TLR) 4 and its antagonist CLI-095, but not by siRNA-mediated knockdown of TLR2. Furthermore, the inhibitors for NF-κB (JSH-23) and mitogen-activated protein kinases (MAPKs) p38 (SB203580) and JNK (SP600125) could also attenuate the effects of SAA1, while the inhibitor for MAPK ERK1/2 (PD 98059) could block the effects of SAA1 only on MMP-1, MMP-8, and LOXL1 but not on MMP-13. CONCLUSION These data highlight a possible role for SAA1 in ECM remodeling preceding membrane rupture by regulating the expression of collagenases MMP-1, MMP-8, MMP-13, and LOXL1 through TLR4-mediated activation of the NF-κB and MAPK pathways in amnion fibroblasts.
Collapse
Affiliation(s)
- Ya-Wei Wang
- Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai, China
| | - Wang-Sheng Wang
- Center for Reproductive Medicine, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai, China
| | - Lu-Yao Wang
- Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai, China
| | - Yi-Rong Bao
- Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai, China
| | - Jiang-Wen Lu
- Center for Reproductive Medicine, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai, China
| | - Yi Lu
- Center for Reproductive Medicine, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai, China
| | - Chu-Yue Zhang
- Center for Reproductive Medicine, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai, China
| | - Wen-Jiao Li
- Maternity and Infant Hospital of Changning District, Shanghai, China
| | - Kang Sun
- Center for Reproductive Medicine, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai, China
| | - Hao Ying
- Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai, China
| |
Collapse
|
19
|
Li L, Zhu H, Zuo X. Interleukin-33 in Systemic Sclerosis: Expression and Pathogenesis. Front Immunol 2018; 9:2663. [PMID: 30498500 PMCID: PMC6249369 DOI: 10.3389/fimmu.2018.02663] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2018] [Accepted: 10/29/2018] [Indexed: 01/05/2023] Open
Abstract
Interleukin-33 (IL-33), a member of the IL-1 superfamily, functions as a traditional cytokine and nuclear factor. It is proposed to have an “alarmin” role. IL-33 mediates biological effects by interacting with the ST2 receptor and IL-1 receptor accessory protein, particularly in innate immune cells and T helper 2 cells. Recent articles have described IL-33 as an emerging pro-fibrotic cytokine in the immune system as well as a novel potential target for systemic sclerosis. Here, we review the available information and focus on the pleiotropic expression and pathogenesis of IL-33 in systemic sclerosis, as well as the feasibility of using IL-33 in clinical applications.
Collapse
Affiliation(s)
- Liya Li
- Department of Rheumatology and immunology, Xiangya Hospital, Central South University, Changsha, China.,The Institute of Rheumatology and Immunology, Central South University, Changsha, China
| | - Honglin Zhu
- Department of Rheumatology and immunology, Xiangya Hospital, Central South University, Changsha, China.,The Institute of Rheumatology and Immunology, Central South University, Changsha, China
| | - Xiaoxia Zuo
- Department of Rheumatology and immunology, Xiangya Hospital, Central South University, Changsha, China.,The Institute of Rheumatology and Immunology, Central South University, Changsha, China
| |
Collapse
|
20
|
Burgess EJ, Hoyt LR, Randall MJ, Mank MM, Bivona JJ, Eisenhauer PL, Botten JW, Ballif BA, Lam YW, Wargo MJ, Boyson JE, Ather JL, Poynter ME. Bacterial Lipoproteins Constitute the TLR2-Stimulating Activity of Serum Amyloid A. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2018; 201:2377-2384. [PMID: 30158125 PMCID: PMC6179936 DOI: 10.4049/jimmunol.1800503] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/11/2018] [Accepted: 08/04/2018] [Indexed: 12/21/2022]
Abstract
Studies comparing endogenous and recombinant serum amyloid A (SAA) have generated conflicting data on the proinflammatory function of these proteins. In exploring this discrepancy, we found that in contrast to commercially sourced recombinant human SAA1 (hSAA1) proteins produced in Escherichia coli, hSAA1 produced from eukaryotic cells did not promote proinflammatory cytokine production from human or mouse cells, induce Th17 differentiation, or stimulate TLR2. Proteomic analysis of E. coli-derived hSAA1 revealed the presence of numerous bacterial proteins, with several being reported or probable lipoproteins. Treatment of hSAA1 with lipoprotein lipase or addition of a lipopeptide to eukaryotic cell-derived hSAA1 inhibited or induced the production of TNF-α from macrophages, respectively. Our results suggest that a function of SAA is in the binding of TLR2-stimulating bacterial proteins, including lipoproteins, and demand that future studies of SAA employ a recombinant protein derived from eukaryotic cells.
Collapse
Affiliation(s)
- Edward J Burgess
- Vermont Lung Center, University of Vermont, Burlington, VT 05405
- Cellular, Molecular, and Biomedical Sciences Program, University of Vermont, Burlington, VT 05405
- Division of Pulmonary Disease and Critical Care, Department of Medicine, University of Vermont, Burlington, VT 05405
| | - Laura R Hoyt
- Vermont Lung Center, University of Vermont, Burlington, VT 05405
- Division of Pulmonary Disease and Critical Care, Department of Medicine, University of Vermont, Burlington, VT 05405
| | - Matthew J Randall
- Vermont Lung Center, University of Vermont, Burlington, VT 05405
- Division of Pulmonary Disease and Critical Care, Department of Medicine, University of Vermont, Burlington, VT 05405
| | - Madeleine M Mank
- Vermont Lung Center, University of Vermont, Burlington, VT 05405
- Division of Pulmonary Disease and Critical Care, Department of Medicine, University of Vermont, Burlington, VT 05405
| | - Joseph J Bivona
- Vermont Lung Center, University of Vermont, Burlington, VT 05405
- Cellular, Molecular, and Biomedical Sciences Program, University of Vermont, Burlington, VT 05405
- Division of Pulmonary Disease and Critical Care, Department of Medicine, University of Vermont, Burlington, VT 05405
| | - Philip L Eisenhauer
- Immunobiology Division, Department of Medicine, University of Vermont, Burlington, VT 05405
| | - Jason W Botten
- Vermont Lung Center, University of Vermont, Burlington, VT 05405
- Cellular, Molecular, and Biomedical Sciences Program, University of Vermont, Burlington, VT 05405
- Immunobiology Division, Department of Medicine, University of Vermont, Burlington, VT 05405
- Department of Microbiology and Molecular Genetics, University of Vermont, Burlington, VT 05405
| | - Bryan A Ballif
- Department of Biology, University of Vermont, Burlington, VT 05405; and
| | - Ying-Wai Lam
- Department of Biology, University of Vermont, Burlington, VT 05405; and
| | - Matthew J Wargo
- Vermont Lung Center, University of Vermont, Burlington, VT 05405
- Cellular, Molecular, and Biomedical Sciences Program, University of Vermont, Burlington, VT 05405
- Department of Microbiology and Molecular Genetics, University of Vermont, Burlington, VT 05405
| | - Jonathan E Boyson
- Vermont Lung Center, University of Vermont, Burlington, VT 05405
- Cellular, Molecular, and Biomedical Sciences Program, University of Vermont, Burlington, VT 05405
- Department of Surgery, University of Vermont, Burlington, VT 05405
| | - Jennifer L Ather
- Vermont Lung Center, University of Vermont, Burlington, VT 05405
- Division of Pulmonary Disease and Critical Care, Department of Medicine, University of Vermont, Burlington, VT 05405
| | - Matthew E Poynter
- Vermont Lung Center, University of Vermont, Burlington, VT 05405;
- Cellular, Molecular, and Biomedical Sciences Program, University of Vermont, Burlington, VT 05405
- Division of Pulmonary Disease and Critical Care, Department of Medicine, University of Vermont, Burlington, VT 05405
| |
Collapse
|
21
|
Wang D, Gao S, Chen J, Zhao Y, Jiang Y, Chu X, Wang X, Liu N, Qin T, Yi Q, Yue Y, Wang S. Dectin-1 stimulates IL-33 expression in dendritic cells via upregulation of IRF4. J Transl Med 2018; 98:708-714. [PMID: 29540860 DOI: 10.1038/s41374-018-0047-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2017] [Revised: 01/11/2018] [Accepted: 01/24/2018] [Indexed: 12/25/2022] Open
Abstract
Interleukin-33 (IL-33) is a potent contributor to antiviral immune responses and antitumor immunity. We recently discovered that IL-33 is overexpressed in dectin-1-activated dendritic cells (DCs). However, mechanisms of dectin-1-induced IL-33 expression in DCs remain elusive. Curdlan, an agonist of dectin-1, was used to mature DCs in this study. We found that dectin-1-induced IL-33 expression in DCs relies on Syk and Raf-1 pathways. By using nuclear factor (NF)-κB inhibitors, we also found that dectin-1-induced IL-33 expression relies on NF-κB signaling. Furthermore, through Syk/Raf-1-NF-κB pathway, dectin-1 signaling stimulates DCs to overexpress interferon regulatory factor 4 (IRF4), which directly upregulates the expression of IL-33 in dectin-1-activated DCs. Thus, our study provides new insights into the mechanisms of dectin-1-induced IL-33 expression in DCs and may provide new targets for improving DC-based cancer immunotherapy.
Collapse
Affiliation(s)
- Dongjiao Wang
- Department of Gynecological Oncology, The First Hospital of Jilin University, Changchun, 130061, China
| | - Sujun Gao
- Department of Hematology, The First Hospital of Jilin University, Changchun, 130061, China
| | - Jintong Chen
- Department of Cancer Immunology, The First Hospital of Jilin University, Changchun, 130061, China
| | - Yinghua Zhao
- Department of Cancer Immunology, The First Hospital of Jilin University, Changchun, 130061, China
| | - Yuxue Jiang
- Department of Cancer Immunology, The First Hospital of Jilin University, Changchun, 130061, China
| | - Xiao Chu
- Department of Cancer Immunology, The First Hospital of Jilin University, Changchun, 130061, China
| | - Xiaohua Wang
- Department of Internal Medicine, Linhai First People's Hospital, Linhai, Zhejiang, 317000, China
| | - Ning Liu
- Department of Gynecological Oncology, The First Hospital of Jilin University, Changchun, 130061, China
| | - Tianxue Qin
- Department of Hematology, The First Hospital of Jilin University, Changchun, 130061, China
| | - Qing Yi
- Department of Cancer Immunology, The First Hospital of Jilin University, Changchun, 130061, China.,Department of Cancer Biology, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, 44195, USA
| | - Ying Yue
- Department of Gynecological Oncology, The First Hospital of Jilin University, Changchun, 130061, China.
| | - Siqing Wang
- Department of Cancer Immunology, The First Hospital of Jilin University, Changchun, 130061, China.
| |
Collapse
|
22
|
Serrano I, Luque A, Aran JM. Exploring the Immunomodulatory Moonlighting Activities of Acute Phase Proteins for Tolerogenic Dendritic Cell Generation. Front Immunol 2018; 9:892. [PMID: 29760704 PMCID: PMC5936965 DOI: 10.3389/fimmu.2018.00892] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2017] [Accepted: 04/10/2018] [Indexed: 12/20/2022] Open
Abstract
The acute phase response is generated by an overwhelming immune-inflammatory process against infection or tissue damage, and represents the initial response of the organism in an attempt to return to homeostasis. It is mediated by acute phase proteins (APPs), an assortment of highly conserved plasma reactants of seemingly different functions that, however, share a common protective role from injury. Recent studies have suggested a crosstalk between several APPs and the mononuclear phagocyte system (MPS) in the resolution of inflammation, to restore tissue integrity and function. In fact, monocyte-derived dendritic cells (Mo-DCs), an integral component of the MPS, play a fundamental role both in the regulation of antigen-specific adaptive responses and in the development of immunologic memory and tolerance, particularly in inflammatory settings. Due to their high plasticity, Mo-DCs can be modeled in vitro toward a tolerogenic phenotype for the treatment of aberrant immune-inflammatory conditions such as autoimmune diseases and allotransplantation, with the phenotypic outcome of these cells depending on the immunomodulatory agent employed. Yet, recent immunotherapy trials have emphasized the drawbacks and challenges facing tolerogenic Mo-DC generation for clinical use, such as reduced therapeutic efficacy and limited in vivo stability of the tolerogenic activity. In this review, we will underline the potential relevance and advantages of APPs for tolerogenic DC production with respect to currently employed immunomodulatory/immunosuppressant compounds. A further understanding of the mechanisms of action underlying the moonlighting immunomodulatory activities exhibited by several APPs over DCs could lead to more efficacious, safe, and stable protocols for precision tolerogenic immunotherapy.
Collapse
Affiliation(s)
- Inmaculada Serrano
- Immune-Inflammatory Processes and Gene Therapeutics Group, Institut d'Investigació Biomèdica de Bellvitge (IDIBELL), L'Hospitalet de Llobregat, Barcelona, Spain
| | - Ana Luque
- Immune-Inflammatory Processes and Gene Therapeutics Group, Institut d'Investigació Biomèdica de Bellvitge (IDIBELL), L'Hospitalet de Llobregat, Barcelona, Spain
| | - Josep M Aran
- Immune-Inflammatory Processes and Gene Therapeutics Group, Institut d'Investigació Biomèdica de Bellvitge (IDIBELL), L'Hospitalet de Llobregat, Barcelona, Spain
| |
Collapse
|
23
|
Gates-Tanzer LT, Shisler JL. Cellular FLIP long isoform (cFLIP L)-IKKα interactions inhibit IRF7 activation, representing a new cellular strategy to inhibit IFNα expression. J Biol Chem 2018; 293:1745-1755. [PMID: 29222334 PMCID: PMC5798304 DOI: 10.1074/jbc.ra117.000541] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2017] [Revised: 11/16/2017] [Indexed: 01/01/2023] Open
Abstract
Interferon α (IFNα) is important for antiviral and anticancer defenses. However, overproduction is associated with autoimmune disorders. Thus, the cell must precisely up- and down-regulate IFNα to achieve immune system homeostasis. The cellular FLICE-like inhibitory protein (cFLIP) is reported to inhibit IFNα production. However, the mechanism for this antagonism remained unknown. The goal here was to identify this mechanism. Here we examined the signal transduction events that occur during TLR9-induced IRF7 activation. The cFLIP long isoform (cFLIPL) inhibited the expression of IRF7-controlled natural or synthetic genes in several cell lines, including those with abundant IRF7 protein levels (e.g. dendritic cells). cFLIPL inhibited IRF7 phosphorylation; however, cFLIPL-IRF7 interactions were not detectable, implying that cFLIPL acted upstream of IRF7 dimerization. Interestingly, cFLIPL co-immunoprecipitated with IKKα, and these interactions correlated with a loss of IKKα-IRF7 interactions. Thus, cFLIP appears to bind to IKKα to prevent IKKα from phosphorylating and activating IRF7. To the best of our knowledge, this is the first report of a cellular protein that uses this approach to inhibit IRF7 activation. Perhaps this cFLIP property could be engineered to minimize the deleterious effects of IFNα expression that occur during certain autoimmune disorders.
Collapse
Affiliation(s)
| | - Joanna L Shisler
- From the Department of Microbiology, University of Illinois, Urbana, Illinois 61801
| |
Collapse
|
24
|
Zhou H, Chen M, Zhang G, Ye RD. Suppression of Lipopolysaccharide-Induced Inflammatory Response by Fragments from Serum Amyloid A. THE JOURNAL OF IMMUNOLOGY 2017; 199:1105-1112. [PMID: 28674180 DOI: 10.4049/jimmunol.1700470] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/30/2017] [Accepted: 06/06/2017] [Indexed: 11/19/2022]
Abstract
Serum amyloid A (SAA) is known as an acute-phase protein and a biomarker for inflammatory diseases. Published studies have shown that SAA possesses proinflammatory cytokine-like activity and is chemotactic for phagocytes, but the structural basis for these activities remains unidentified. In this article, we report that truncated SAA1 proteins lacking N- and C-terminal sequences exhibit reduced proinflammatory activity and strongly suppress LPS-induced expression of IL-1β, IL-6, and TNF-α in macrophages. A truncated SAA1 containing aa 11-58 was examined further and found to facilitate p38 MAPK phosphorylation while reducing LPS-stimulated phosphorylation of ERK and JNK. In LPS-challenged mice, aa 11-58 reduced the severity of acute lung injury, with significantly less neutrophil infiltration in the lungs and attenuated pulmonary expression of IL-1β, IL-6, and TNF-α. Coadministration of aa 11-58 markedly improved mouse survival in response to a lethal dose of LPS. A potent induction of IL-10 was observed in a TLR2-dependent, but TLR4-independent, manner in macrophages stimulated with aa 11-58. However, the aa 11-58 fragment of SAA1 was unable to induce chemotaxis or calcium flux through formyl peptide receptor 2. These results indicate that the N- and C-terminal sequences contain structural determinants for the proinflammatory and chemotactic activities of SAA1, and their removal switches SAA1 to an anti-inflammatory role. Given that proteolytic processing of SAA is associated with the pathological changes in several diseases, including secondary amyloidosis, our findings may shed light on the structure-function relationship of SAA1 with respect to its role in inflammation.
Collapse
Affiliation(s)
- Huibin Zhou
- Engineering Research Center of Cell and Therapeutic Antibody, Ministry of Education, School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, China; and
| | - Mingjie Chen
- Engineering Research Center of Cell and Therapeutic Antibody, Ministry of Education, School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, China; and
| | - Gufang Zhang
- Engineering Research Center of Cell and Therapeutic Antibody, Ministry of Education, School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, China; and
| | - Richard D Ye
- Engineering Research Center of Cell and Therapeutic Antibody, Ministry of Education, School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, China; and .,Institute of Chinese Medical Sciences, University of Macau, Macau Special Administrative Region 999078, China
| |
Collapse
|
25
|
Watanabe T, Yamashita K, Arai Y, Minaga K, Kamata K, Nagai T, Komeda Y, Takenaka M, Hagiwara S, Ida H, Sakurai T, Nishida N, Strober W, Kudo M. Chronic Fibro-Inflammatory Responses in Autoimmune Pancreatitis Depend on IFN-α and IL-33 Produced by Plasmacytoid Dendritic Cells. THE JOURNAL OF IMMUNOLOGY 2017; 198:3886-3896. [PMID: 28373582 DOI: 10.4049/jimmunol.1700060] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/11/2017] [Accepted: 03/08/2017] [Indexed: 12/22/2022]
Abstract
In previous studies, we found that human IgG4-related autoimmune pancreatitis (AIP) and murine AIP are driven by activation of plasmacytoid dendritic cells (pDCs) producing IFN-α. In the present studies we examined additional roles of pDC-related mechanisms in AIP pathogenesis, particularly those responsible for induction of fibrosis. We found that in murine AIP (MRL/Mp mice treated with polyinosinic-polycytidylic acid) not only the pancreatic infiltration of immune cells but also the development of fibrosis were markedly reduced by the depletion of pDCs or blockade of type I IFN signaling; moreover, such treatment was accompanied by a marked reduction of pancreatic expression of IL-33. Conversely, polyinosinic-polycytidylic acid-induced inflamed pancreatic tissue in murine AIP exhibited increased expression of type I IFNs and IL-33 (and downstream IL-33 cytokines such as IL-13 and TGF-β1). pDCs stimulated by type I IFN were the source of the IL-33 because purified populations of these cells isolated from the inflamed pancreas produced a large amount of IL-33 upon activation by TLR9 ligands, and such production was abrogated by the neutralization of type I IFN. The role of IL-33 in murine AIP pathogenesis was surprisingly important because blockade of IL-33 signaling by anti-ST2 Ab attenuated both pancreatic inflammation and accompanying fibrosis. Finally, whereas patients with both conventional pancreatitis and IgG4-related AIP exhibited increased numbers of acinar cells expressing IL-33, only the latter also exhibited pDCs producing this cytokine. These data thus suggest that pDCs producing IFN-α and IL-33 play a pivotal role in the chronic fibro-inflammatory responses underlying murine AIP and human IgG4-related AIP.
Collapse
Affiliation(s)
- Tomohiro Watanabe
- Department of Gastroenterology and Hepatology, Kindai University Faculty of Medicine, Osaka-Sayama, Osaka 589-8511, Japan; .,Mucosal Immunity Section, Laboratory of Host Defenses, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892; and
| | - Kouhei Yamashita
- Department of Hematology and Oncology, Kyoto University Graduate School of Medicine, Kyoto 606-8507, Japan
| | - Yasuyuki Arai
- Department of Hematology and Oncology, Kyoto University Graduate School of Medicine, Kyoto 606-8507, Japan
| | - Kosuke Minaga
- Department of Gastroenterology and Hepatology, Kindai University Faculty of Medicine, Osaka-Sayama, Osaka 589-8511, Japan
| | - Ken Kamata
- Department of Gastroenterology and Hepatology, Kindai University Faculty of Medicine, Osaka-Sayama, Osaka 589-8511, Japan
| | - Tomoyuki Nagai
- Department of Gastroenterology and Hepatology, Kindai University Faculty of Medicine, Osaka-Sayama, Osaka 589-8511, Japan
| | - Yoriaki Komeda
- Department of Gastroenterology and Hepatology, Kindai University Faculty of Medicine, Osaka-Sayama, Osaka 589-8511, Japan
| | - Mamoru Takenaka
- Department of Gastroenterology and Hepatology, Kindai University Faculty of Medicine, Osaka-Sayama, Osaka 589-8511, Japan
| | - Satoru Hagiwara
- Department of Gastroenterology and Hepatology, Kindai University Faculty of Medicine, Osaka-Sayama, Osaka 589-8511, Japan
| | - Hiroshi Ida
- Department of Gastroenterology and Hepatology, Kindai University Faculty of Medicine, Osaka-Sayama, Osaka 589-8511, Japan
| | - Toshiharu Sakurai
- Department of Gastroenterology and Hepatology, Kindai University Faculty of Medicine, Osaka-Sayama, Osaka 589-8511, Japan
| | - Naoshi Nishida
- Department of Gastroenterology and Hepatology, Kindai University Faculty of Medicine, Osaka-Sayama, Osaka 589-8511, Japan
| | - Warren Strober
- Mucosal Immunity Section, Laboratory of Host Defenses, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892; and
| | - Masatoshi Kudo
- Department of Gastroenterology and Hepatology, Kindai University Faculty of Medicine, Osaka-Sayama, Osaka 589-8511, Japan
| |
Collapse
|
26
|
Stav-Noraas TE, Edelmann RJ, Poulsen LLC, Sundnes O, Phung D, Küchler AM, Müller F, Kamen AA, Haraldsen G, Kaarbø M, Hol J. Endothelial IL-33 Expression Is Augmented by Adenoviral Activation of the DNA Damage Machinery. THE JOURNAL OF IMMUNOLOGY 2017; 198:3318-3325. [PMID: 28258201 DOI: 10.4049/jimmunol.1600054] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/12/2016] [Accepted: 02/06/2017] [Indexed: 01/16/2023]
Abstract
IL-33, required for viral clearance by cytotoxic T cells, is generally expressed in vascular endothelial cells in healthy human tissues. We discovered that endothelial IL-33 expression was stimulated as a response to adenoviral transduction. This response was dependent on MRE11, a sensor of DNA damage that can also be activated by adenoviral DNA, and on IRF1, a transcriptional regulator of cellular responses to viral invasion and DNA damage. Accordingly, we observed that endothelial cells responded to adenoviral DNA by phosphorylation of ATM and CHK2 and that depletion or inhibition of MRE11, but not depletion of ATM, abrogated IL-33 stimulation. In conclusion, we show that adenoviral transduction stimulates IL-33 expression in endothelial cells in a manner that is dependent on the DNA-binding protein MRE11 and the antiviral factor IRF1 but not on downstream DNA damage response signaling.
Collapse
Affiliation(s)
- Tor Espen Stav-Noraas
- K.G. Jebsen Inflammation Research Centre, Oslo University Hospital and University of Oslo, N-0424 Oslo, Norway.,Laboratory of Immunohistochemistry and Immunopathology, Department of Pathology, Oslo University Hospital and University of Oslo, N-0424 Oslo, Norway
| | - Reidunn J Edelmann
- K.G. Jebsen Inflammation Research Centre, Oslo University Hospital and University of Oslo, N-0424 Oslo, Norway.,Laboratory of Immunohistochemistry and Immunopathology, Department of Pathology, Oslo University Hospital and University of Oslo, N-0424 Oslo, Norway
| | - Lars La Cour Poulsen
- K.G. Jebsen Inflammation Research Centre, Oslo University Hospital and University of Oslo, N-0424 Oslo, Norway.,Laboratory of Immunohistochemistry and Immunopathology, Department of Pathology, Oslo University Hospital and University of Oslo, N-0424 Oslo, Norway
| | - Olav Sundnes
- K.G. Jebsen Inflammation Research Centre, Oslo University Hospital and University of Oslo, N-0424 Oslo, Norway.,Laboratory of Immunohistochemistry and Immunopathology, Department of Pathology, Oslo University Hospital and University of Oslo, N-0424 Oslo, Norway
| | - Danh Phung
- Laboratory of Immunohistochemistry and Immunopathology, Department of Pathology, Oslo University Hospital and University of Oslo, N-0424 Oslo, Norway
| | - Axel M Küchler
- Laboratory of Immunohistochemistry and Immunopathology, Department of Pathology, Oslo University Hospital and University of Oslo, N-0424 Oslo, Norway
| | - Fredrik Müller
- Department of Microbiology, Oslo University Hospital and University of Oslo, N-0424 Oslo, Norway; and
| | - Amine A Kamen
- Department of Bioengineering, McGill University, Montreal, Quebec H3A OC3, Canada
| | - Guttorm Haraldsen
- K.G. Jebsen Inflammation Research Centre, Oslo University Hospital and University of Oslo, N-0424 Oslo, Norway; .,Laboratory of Immunohistochemistry and Immunopathology, Department of Pathology, Oslo University Hospital and University of Oslo, N-0424 Oslo, Norway
| | - Mari Kaarbø
- Department of Microbiology, Oslo University Hospital and University of Oslo, N-0424 Oslo, Norway; and
| | - Johanna Hol
- K.G. Jebsen Inflammation Research Centre, Oslo University Hospital and University of Oslo, N-0424 Oslo, Norway.,Laboratory of Immunohistochemistry and Immunopathology, Department of Pathology, Oslo University Hospital and University of Oslo, N-0424 Oslo, Norway
| |
Collapse
|
27
|
Abstract
The innate immune system represents the first line of defense against pathogens and comprises both a cellular and a humoral arm. Fluid-phase pattern recognition molecules (PRMs), which include collectins, ficolins, and pentraxins, are key components of the humoral arm of innate immunity and are expressed by a variety of cells, including myeloid, epithelial, and endothelial cells, mainly in response to infectious and inflammatory conditions. Soluble PRMs share basic multifunctional properties including activation and regulation of the complement cascade, opsonization of pathogens and apoptotic cells, regulation of leukocyte extravasation, and fine-tuning of inflammation. Therefore, soluble PRMs are part of the immune response and retain antibody-like effector functions. Here, we will review the expression and general function of soluble PRMs, focusing our attention on the long pentraxin PTX3.
Collapse
|
28
|
Schrödl W, Büchler R, Wendler S, Reinhold P, Muckova P, Reindl J, Rhode H. Acute phase proteins as promising biomarkers: Perspectives and limitations for human and veterinary medicine. Proteomics Clin Appl 2016; 10:1077-1092. [PMID: 27274000 DOI: 10.1002/prca.201600028] [Citation(s) in RCA: 66] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2016] [Revised: 05/09/2016] [Accepted: 06/01/2016] [Indexed: 12/23/2022]
Abstract
Acute phase proteins (APPs) are highly conserved plasma proteins that are increasingly secreted by the liver in response to a variety of injuries, independently of their location and cause. APPs favor the systemic regulation of defense, coagulation, proteolysis, and tissue repair. Various APPs have been applied as general diagnostic parameters for a long time. Through proteomic techniques, more and more APPs have been discovered to be differentially altered. Since they are not consistently explainable by a stereotypic hepatic expression of sets of APPs, most of these results have unfortunately been neglected or attributed to the nonspecificity of the acute phase reaction. Moreover, it appears that various extrahepatic tissues are also able to express APPs. These extrahepatic APPs show focally specific roles in tissue homeostasis and repair and are released primarily into interstitial and distal fluids. Since these focal proteins might leak into the circulatory system, mixtures of hepatic and extrahepatic APP species can be expected in blood. Hence, a selective alteration of parts of APPs might be expected. There are several hints on multiple molecular forms and fragments of tissue-derived APPs. These differences offer the chance for multiple selective determinations. Thus, specific proteoforms might indeed serve as tissue-specific disease indicators.
Collapse
Affiliation(s)
- Wieland Schrödl
- Institute of Bacteriology and Mycology, Veterinary Faculty, University Leipzig, Germany
| | - Rita Büchler
- Institute of Biochemistry I, University Hospital Jena, Germany
| | - Sindy Wendler
- Institute of Biochemistry I, University Hospital Jena, Germany
| | - Petra Reinhold
- Institute of Molecular Pathogenesis at 'Friedrich Loeffler Institut', Federal Research Institute for Animal Health, Jena, Germany
| | - Petra Muckova
- Institute of Biochemistry I, University Hospital Jena, Germany.,Clinic of Neurology, University Hospital Jena, Germany
| | - Johanna Reindl
- Institute of Biochemistry I, University Hospital Jena, Germany
| | - Heidrun Rhode
- Institute of Biochemistry I, University Hospital Jena, Germany
| |
Collapse
|
29
|
Lu B, Yang M, Wang Q. Interleukin-33 in tumorigenesis, tumor immune evasion, and cancer immunotherapy. J Mol Med (Berl) 2016; 94:535-43. [PMID: 26922618 DOI: 10.1007/s00109-016-1397-0] [Citation(s) in RCA: 66] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2015] [Revised: 02/14/2016] [Accepted: 02/17/2016] [Indexed: 12/20/2022]
Abstract
Interleukin-33 (IL-33) is a member of the IL-1 gene family and mainly expressed in the nucleus of tissue lining cells, stromal cells, and activated myeloid cells. IL-33 is considered a damage-associated molecular pattern (DAMP) molecule and plays an important role in many physiological and pathological settings such as tissue repair, allergy, autoimmune disease, infectious disease, and cancer. The biological functions of IL-33 include maintaining tissue homeostasis, enhancing type 1 and 2 cellular immune responses, and mediating fibrosis during chronic inflammation. IL-33 exerts diverse functions through signaling via its receptor ST2, which is expressed in many types of cells including regulatory T cells (Treg), group 2 innate lymphoid cells (ILC2s), myeloid cells, cytotoxic NK cells, Th2 cells, Th1 cells, and CD8(+) T cells. Tumor development results in downregulation of IL-33 in epithelial cells but upregulation of IL-33 in the tumor stroma and serum. The current data suggest that IL-33 expression in tumor cells increases immunogenicity and promotes type 1 antitumor immune responses through CD8(+) T cells and NK cells, whereas IL-33 in tumor stroma and serum facilitates immune suppression via Treg and myeloid-derived suppressor cell (MDSC). Understanding the role of IL-33 in cancer immunobiology sheds lights on targeting this cytokine for cancer immunotherapy.
Collapse
Affiliation(s)
- Binfeng Lu
- Department of Immunology, University of Pittsburgh School of Medicine, 200 Lothrop Street, Pittsburgh, PA, 15261, USA.
- University of Pittsburgh Cancer Institute, Pittsburgh, PA, USA.
| | - Min Yang
- Department of Immunology, Institute of Medical Biotechnology, School of Biology and Basic Medical Sciences, Soochow University, Suzhou, People's Republic of China
| | - Qingqing Wang
- Institute of Immunology, Zhejiang University School of Medicine, Hangzhou, 310058, China
| |
Collapse
|
30
|
Bosco A, Wiehler S, Proud D. Interferon regulatory factor 7 regulates airway epithelial cell responses to human rhinovirus infection. BMC Genomics 2016; 17:76. [PMID: 26810609 PMCID: PMC4727386 DOI: 10.1186/s12864-016-2405-z] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2015] [Accepted: 01/18/2016] [Indexed: 12/11/2022] Open
Abstract
Background Human rhinoviruses (HRV) cause the majority of colds and trigger exacerbations of chronic lower airway diseases. Airway epithelial cells are the primary site for HRV infection and replication, and the initiation of host inflammatory responses. At present, the molecular mechanisms that underpin HRV responses in airway epithelial cells are incompletely understood. The aim of this study was to employ microarray profiling, upstream regulator analysis, and siRNA mediated gene silencing to further our understanding of the role of interferon regulatory factor 7 (IRF7) in this response. Methods Primary human bronchial epithelial cells (HBE) where transfected with siRNA that targets IRF7 or a non-silencing control (all-star control) using Lipofectamine. The cells were allowed to recover, and then cultured in the presence or absence of HRV-16 for 24 h. Global patterns of gene expression were profiled on microarrays. A subset of genes identified in the microarray study were validated at the mRNA and/or protein level using real time RT-qPCR, ELISA, and western blots. Results Hundreds of genes were upregulated in HBE during HRV infection. Pathways analysis demonstrated that these genes were mainly involved in type I and II interferon signaling, RIG-I/MDA5 signaling, antigen processing and presentation, and apoptosis. Upstream regulator analysis of these data suggested that IRF7 was a major molecular driver of this response. Knockdown of IRF7 reduced the HRV-driven upregulation of genes involved in antiviral responses (interferon signaling, Toll-like receptor signaling, NOD-like receptor signaling, RIG-I/MDA5 signaling), and increased the expression of genes that promote inflammation (e.g. CXCL5, IL-33, IL1RL1) and the response to oxidative stress. However, the majority of genes that were perturbed by HRV in HBE cells including those that are known to be regulated by IRF7 were insensitive to IRF7 knockdown. Upstream regulator analysis of the part of the response that was insensitive to IRF7 knockdown suggested it was driven by NF-κB, STAT1, STAT3, and IRF1. Conclusions Our findings demonstrate that IRF7 regulates the expression of genes involved in antiviral immunity, inflammation, and the response to oxidative stress during HRV infections in HBE cells, and also suggests that other transcription factors play a major role in this response. Electronic supplementary material The online version of this article (doi:10.1186/s12864-016-2405-z) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Anthony Bosco
- Telethon Kids Institute, University of Western Australia, PO Box 855, West Perth, WA, 6872, Australia.
| | - Shahina Wiehler
- Airway Inflammation Research Group, Snyder Institute for Chronic Diseases, and the Department of Physiology & Pharmacology, University of Calgary Faculty of Medicine, Calgary, AB, Canada.
| | - David Proud
- Airway Inflammation Research Group, Snyder Institute for Chronic Diseases, and the Department of Physiology & Pharmacology, University of Calgary Faculty of Medicine, Calgary, AB, Canada.
| |
Collapse
|
31
|
De Buck M, Gouwy M, Wang JM, Van Snick J, Proost P, Struyf S, Van Damme J. The cytokine-serum amyloid A-chemokine network. Cytokine Growth Factor Rev 2015; 30:55-69. [PMID: 26794452 DOI: 10.1016/j.cytogfr.2015.12.010] [Citation(s) in RCA: 99] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2015] [Revised: 12/22/2015] [Accepted: 12/22/2015] [Indexed: 12/14/2022]
Abstract
Levels of serum amyloid A (SAA), a major acute phase protein in humans, are increased up to 1000-fold upon infection, trauma, cancer or other inflammatory events. However, the exact role of SAA in host defense is yet not fully understood. Several pro- and anti-inflammatory properties have been ascribed to SAA. Here, the regulated production of SAA by cytokines and glucocorticoids is discussed first. Secondly, the cytokine and chemokine inducing capacity of SAA and its receptor usage are reviewed. Thirdly, the direct (via FPR2) and indirect (via TLR2) chemotactic effects of SAA and its synergy with chemokines are unraveled. Altogether, a complex cytokine-SAA-chemokine network is established, in which SAA plays a key role in regulating the inflammatory response.
Collapse
Affiliation(s)
- Mieke De Buck
- KU Leuven, University of Leuven, Department of Microbiology and Immunology, Rega Institute for Medical Research, Laboratory of Molecular Immunology, Minderbroedersstraat 10, 3000 Leuven, Belgium.
| | - Mieke Gouwy
- KU Leuven, University of Leuven, Department of Microbiology and Immunology, Rega Institute for Medical Research, Laboratory of Molecular Immunology, Minderbroedersstraat 10, 3000 Leuven, Belgium.
| | - Ji Ming Wang
- Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute at Frederick, Frederick, MD 21702, USA.
| | - Jacques Van Snick
- Ludwig Cancer Research, Brussels Branch, Brussels, Belgium; e Duve Institute, Université Catholique de Louvain, Brussels, Belgium.
| | - Paul Proost
- KU Leuven, University of Leuven, Department of Microbiology and Immunology, Rega Institute for Medical Research, Laboratory of Molecular Immunology, Minderbroedersstraat 10, 3000 Leuven, Belgium.
| | - Sofie Struyf
- KU Leuven, University of Leuven, Department of Microbiology and Immunology, Rega Institute for Medical Research, Laboratory of Molecular Immunology, Minderbroedersstraat 10, 3000 Leuven, Belgium.
| | - Jo Van Damme
- KU Leuven, University of Leuven, Department of Microbiology and Immunology, Rega Institute for Medical Research, Laboratory of Molecular Immunology, Minderbroedersstraat 10, 3000 Leuven, Belgium.
| |
Collapse
|
32
|
Molofsky AB, Savage AK, Locksley RM. Interleukin-33 in Tissue Homeostasis, Injury, and Inflammation. Immunity 2015; 42:1005-19. [PMID: 26084021 DOI: 10.1016/j.immuni.2015.06.006] [Citation(s) in RCA: 487] [Impact Index Per Article: 48.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2015] [Indexed: 12/12/2022]
Abstract
Interleukin-33 (IL-33) is a nuclear-associated cytokine of the IL-1 family originally described as a potent inducer of allergic type 2 immunity. IL-33 signals via the receptor ST2, which is highly expressed on group 2 innate lymphoid cells (ILC2s) and T helper 2 (Th2) cells, thus underpinning its association with helminth infection and allergic pathology. Recent studies have revealed ST2 expression on subsets of regulatory T cells, and for a role for IL-33 in tissue homeostasis and repair that suggests previously unrecognized interactions within these cellular networks. IL-33 can participate in pathologic fibrotic reactions, or, in the setting of microbial invasion, can cooperate with inflammatory cytokines to promote responses by cytotoxic NK cells, Th1 cells, and CD8(+) T cells. Here, we highlight the regulation and function of IL-33 and ST2 and review their roles in homeostasis, damage, and inflammation, suggesting a conceptual framework for future studies.
Collapse
Affiliation(s)
- Ari B Molofsky
- Department of Microbiology & Immunology, University of California, San Francisco, 94143-0795, USA; Department of Laboratory Medicine, University of California, San Francisco, 94143-0795, USA
| | - Adam K Savage
- Howard Hughes Medical Institute, University of California, San Francisco, 94143-0795, USA; Department of Microbiology & Immunology, University of California, San Francisco, 94143-0795, USA
| | - Richard M Locksley
- Howard Hughes Medical Institute, University of California, San Francisco, 94143-0795, USA; Department of Medicine, University of California, San Francisco, 94143-0795, USA; Department of Microbiology & Immunology, University of California, San Francisco, 94143-0795, USA.
| |
Collapse
|
33
|
De Buck M, Berghmans N, Pörtner N, Vanbrabant L, Cockx M, Struyf S, Opdenakker G, Proost P, Van Damme J, Gouwy M. Serum amyloid A1α induces paracrine IL-8/CXCL8 via TLR2 and directly synergizes with this chemokine via CXCR2 and formyl peptide receptor 2 to recruit neutrophils. J Leukoc Biol 2015; 98:1049-60. [PMID: 26297794 DOI: 10.1189/jlb.3a0315-085r] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2015] [Accepted: 07/23/2015] [Indexed: 11/24/2022] Open
Abstract
Cell migration depends on the ability of leukocytes to sense an external gradient of chemotactic proteins produced during inflammation. These proteins include chemokines, complement factors, and some acute phase proteins, such as serum amyloid A. Serum amyloid A chemoattracts neutrophils, monocytes, and T lymphocytes via its G protein-coupled receptor formyl peptide receptor 2. We demonstrate that serum amyloid A1α more potently chemoattracts neutrophils in vivo than in vitro. In contrast to CD14(+) monocytes, no rapid (within 2 h) induction of interleukin-8/CXC chemokine ligand 8 or macrophage-inflammatory protein-1α/CC chemokine ligand 3 was observed in purified human neutrophils after stimulation of the cells with serum amyloid A1α or lipopolysaccharide. Moreover, interleukin-8/CXC chemokine ligand 8 induction in monocytes by serum amyloid A1α was mediated by toll-like receptor 2 and was inhibited by association of serum amyloid A1α with high density lipoprotein. This indicates that the potent chemotactic response of neutrophils toward intraperitoneally injected serum amyloid A1α is indirectly enhanced by rapid induction of chemokines in peritoneal cells, synergizing in a paracrine manner with serum amyloid A1α. We observed direct synergy between IL-8/CXC chemokine ligand 8 and serum amyloid A1α, but not lipopolysaccharide, in chemotaxis and shape change assays with neutrophils. Furthermore, the selective CXC chemokine receptor 2 and formyl peptide receptor 2 antagonists, SB225002 and WRW4, respectively, blocked the synergy between IL-8/CXC chemokine ligand 8 and serum amyloid A1α in neutrophil chemotaxis in vitro, indicating that for synergy their corresponding G protein-coupled receptors are required. Additionally, SB225002 significantly inhibited serum amyloid A1α-mediated peritoneal neutrophil influx. Taken together, endogenous (e.g., IL-1β) and exogenous (e.g., lipopolysaccharide) inflammatory mediators induce primary chemoattractants such as serum amyloid A that synergize in an autocrine (monocyte) or a paracrine (neutrophil) fashion with secondary chemokines induced in stromal cells.
Collapse
Affiliation(s)
- Mieke De Buck
- *Laboratory of Molecular Immunology and Laboratory of Immunobiology, Rega Institute for Medical Research, Department of Microbiology and Immunology, University of Leuven, Leuven, Belgium
| | - Nele Berghmans
- *Laboratory of Molecular Immunology and Laboratory of Immunobiology, Rega Institute for Medical Research, Department of Microbiology and Immunology, University of Leuven, Leuven, Belgium
| | - Noëmie Pörtner
- *Laboratory of Molecular Immunology and Laboratory of Immunobiology, Rega Institute for Medical Research, Department of Microbiology and Immunology, University of Leuven, Leuven, Belgium
| | - Lotte Vanbrabant
- *Laboratory of Molecular Immunology and Laboratory of Immunobiology, Rega Institute for Medical Research, Department of Microbiology and Immunology, University of Leuven, Leuven, Belgium
| | - Maaike Cockx
- *Laboratory of Molecular Immunology and Laboratory of Immunobiology, Rega Institute for Medical Research, Department of Microbiology and Immunology, University of Leuven, Leuven, Belgium
| | - Sofie Struyf
- *Laboratory of Molecular Immunology and Laboratory of Immunobiology, Rega Institute for Medical Research, Department of Microbiology and Immunology, University of Leuven, Leuven, Belgium
| | - Ghislain Opdenakker
- *Laboratory of Molecular Immunology and Laboratory of Immunobiology, Rega Institute for Medical Research, Department of Microbiology and Immunology, University of Leuven, Leuven, Belgium
| | - Paul Proost
- *Laboratory of Molecular Immunology and Laboratory of Immunobiology, Rega Institute for Medical Research, Department of Microbiology and Immunology, University of Leuven, Leuven, Belgium
| | - Jo Van Damme
- *Laboratory of Molecular Immunology and Laboratory of Immunobiology, Rega Institute for Medical Research, Department of Microbiology and Immunology, University of Leuven, Leuven, Belgium
| | - Mieke Gouwy
- *Laboratory of Molecular Immunology and Laboratory of Immunobiology, Rega Institute for Medical Research, Department of Microbiology and Immunology, University of Leuven, Leuven, Belgium
| |
Collapse
|
34
|
Ye RD, Sun L. Emerging functions of serum amyloid A in inflammation. J Leukoc Biol 2015; 98:923-9. [PMID: 26130702 DOI: 10.1189/jlb.3vmr0315-080r] [Citation(s) in RCA: 215] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2015] [Accepted: 06/02/2015] [Indexed: 12/12/2022] Open
Abstract
SAA is a major acute-phase protein produced in large quantity during APR. The rise of SAA concentration in blood circulation during APR has been a clinical marker for active inflammation. In the past decade, research has been conducted to determine whether SAA plays an active role during inflammation and if so, how it influences the course of inflammation. These efforts have led to the discovery of cytokine-like activities of rhSAA, which is commercially available and widely used in most of the published studies. SAA activates multiple receptors, including the FPR2, the TLRs TLR2 and TLR4, the scavenger receptor SR-BI, and the ATP receptor P2X7. More recent studies have shown that SAA not only activates transcription factors, such as NF-κB, but also plays a role in epigenetic regulation through a MyD88-IRF4-Jmjd3 pathway. It is postulated that the activation of these pathways leads to induced expression of proinflammatory factors and a subset of proteins expressed by the M2 macrophages. These functional properties set SAA apart from well-characterized inflammatory factors, such as LPS and TNF-α, suggesting that it may play a homeostatic role during the course of inflammation. Ongoing and future studies are directed to addressing unresolved issues, including the difference between rSAA and native SAA isoforms and the exact functions of SAA in physiologic and pathologic settings.
Collapse
Affiliation(s)
- Richard D Ye
- *School of Pharmacy, Shanghai Jiao Tong University, Shanghai, China; and Department of Pharmacology, University of Illinois at Chicago, Illinois, USA
| | - Lei Sun
- *School of Pharmacy, Shanghai Jiao Tong University, Shanghai, China; and Department of Pharmacology, University of Illinois at Chicago, Illinois, USA
| |
Collapse
|
35
|
Sozzani S, Del Prete A. Chemokines as relay signals in human dendritic cell migration: serum amyloid A kicks off chemotaxis. Eur J Immunol 2015; 45:40-3. [PMID: 25472709 DOI: 10.1002/eji.201445305] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2014] [Revised: 11/19/2014] [Accepted: 12/02/2014] [Indexed: 12/31/2022]
Abstract
Cell migration is a response highly conserved in evolution. Chemotactic factors secreted in injured and inflamed tissues generate a concentration-based, chemotactic gradient that directs leukocytes from the blood compartment into tissue. In this issue of the European Journal of Immunology, Gouwy et al. [Eur. J. Immunol. 2015. 45: 101-112] show that the SAA1α isoform of serum amyloid A (SAA), which is an acute phase protein upregulated in inflammation and shown to chemoattract some leukocyte subsets, is also able to chemoattract monocyte-derived immature dendritic cells (DCs). The authors also show that the chemotactic activity of SAA1α for monocytes and DCs is indirectly mediated by rapid chemokine induction, providing evidence that proposes a new level of regulation of leukocyte migration.
Collapse
Affiliation(s)
- Silvano Sozzani
- Department of Molecular and Translational Medicine, University of Brescia, Brescia and Humanitas Clinical and Research Institute, Rozzano, Italy
| | | |
Collapse
|
36
|
Chen M, Zhou H, Cheng N, Qian F, Ye RD. Serum amyloid A1 isoforms display different efficacy at Toll-like receptor 2 and formyl peptide receptor 2. Immunobiology 2014; 219:916-23. [PMID: 25154907 PMCID: PMC4252704 DOI: 10.1016/j.imbio.2014.08.002] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2014] [Revised: 07/22/2014] [Accepted: 08/03/2014] [Indexed: 01/13/2023]
Abstract
Serum amyloid A (SAA) is a major acute-phase protein and a precursor of amyloid A, the deposit of which leads to amyloidosis. Different alleles exist in SAA1, a predominant form of the human SAA gene family. Emerging evidence has shown correlations between these alleles and diseases including familiar Mediterranean fever and amyloidosis. However, it remains unclear how the proteins encoded by these SAA1 alleles act differently. Here we report the characterization of proteins encoded by SAA1.1, SAA1.3, and SAA1.5, in comparison to that encoded by SAA2.2, for their preference of the SAA receptors including formyl peptide receptor 2 (FPR2) and Toll-like receptor 2 (TLR2). SAA1.1 was more efficacious than SAA1.3 and SAA1.5 but equally efficacious to SAA2.2 in calcium mobilization and chemotaxis assays, which measure the activation of the G protein-coupled FPR2. In agreement with this, SAA1.1 and SAA2.2 induced more robust phosphorylation of ERK than SAA1.3 and SAA1.5. Only small differences were observed between the SAA1 proteins tested and SAA2.2 in TLR2-dependent NF-κB luciferase reporter assay. In comparison, SAA1.3 was most effective in stimulating ERK and p38 MAPK phosphorylation. Using bone marrow-derived macrophages from C57BL/10ScN (Tlr4lps-del) mice, we examined the SAA isoforms for their induction of selected pro- and anti-inflammatory cytokines. SAA1.3 was most potent in the induction of TNFα and IL-1rn, whereas SAA1.5 induced robust IL-10 expression. These results show differences of the SAA1 isoforms in their selectivity for the SAA receptors, which may affect their roles in modulating inflammation and immunity.
Collapse
Affiliation(s)
- Mingjie Chen
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai, PR China
| | - Huibing Zhou
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai, PR China
| | - Ni Cheng
- Department of Pharmacology, University of Illinois College of Medicine, Chicago, Illinois 60612, USA
| | - Feng Qian
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai, PR China
| | - Richard D Ye
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai, PR China; Department of Pharmacology, University of Illinois College of Medicine, Chicago, Illinois 60612, USA.
| |
Collapse
|
37
|
Gouwy M, De Buck M, Pörtner N, Opdenakker G, Proost P, Struyf S, Van Damme J. Serum amyloid A chemoattracts immature dendritic cells and indirectly provokes monocyte chemotaxis by induction of cooperating CC and CXC chemokines. Eur J Immunol 2014; 45:101-12. [PMID: 25345597 DOI: 10.1002/eji.201444818] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2014] [Revised: 09/11/2014] [Accepted: 10/21/2014] [Indexed: 12/21/2022]
Abstract
Serum amyloid A (SAA) is an acute phase protein that is upregulated in inflammatory diseases and chemoattracts monocytes, lymphocytes, and granulocytes via its G protein-coupled receptor formyl peptide receptor like 1/formyl peptide receptor 2 (FPRL1/FPR2). Here, we demonstrated that the SAA1α isoform also chemoattracts monocyte-derived immature dendritic cells (DCs) in the Boyden and μ-slide chemotaxis assay and that its chemotactic activity for monocytes and DCs was indirectly mediated via rapid chemokine induction. Indeed, SAA1 induced significant amounts (≥5 ng/mL) of macrophage inflammatory protein-1α/CC chemokine ligand 3 (MIP-1α/CCL3) and interleukin-8/CXC chemokine ligand 8 (IL-8/CXCL8) in monocytes and DCs in a dose-dependent manner within 3 h. However, SAA1 also directly activated monocytes and DCs for signaling and chemotaxis without chemokine interference. SAA1-induced monocyte migration was nevertheless significantly prevented (60-80% inhibition) in the constant presence of desensitizing exogenous MIP-1α/CCL3, neutralizing anti-MIP-1α/CCL3 antibody, or a combination of CC chemokine receptor 1 (CCR1) and CCR5 antagonists, indicating that this endogenously produced CC chemokine was indirectly contributing to SAA1-mediated chemotaxis. Further, anti-IL-8/CXCL8 antibody neutralized SAA1-induced monocyte migration, suggesting that endogenous IL-8/CXCL8 acted in concert with MIP-1α/CCL3. This explained why SAA1 failed to synergize with exogenously added MIP-1α/CCL3 or stromal cell-derived factor-1α (SDF-1α)/CXCL12 in monocyte and DC chemotaxis. In addition to direct leukocyte activation, SAA1 induces a chemotactic cascade mediated by expression of cooperating chemokines to prolong leukocyte recruitment to the inflammatory site.
Collapse
Affiliation(s)
- Mieke Gouwy
- Laboratory of Molecular Immunology, Rega Institute for Medical Research, Department of Microbiology and Immunology, University of Leuven, Leuven, Belgium
| | | | | | | | | | | | | |
Collapse
|