1
|
Bataclan M, Leoni C, Moro SG, Pecoraro M, Wong EH, Heissmeyer V, Monticelli S. Crosstalk between Regnase-1 and -3 shapes mast cell survival and cytokine expression. Life Sci Alliance 2024; 7:e202402784. [PMID: 38830770 PMCID: PMC11147952 DOI: 10.26508/lsa.202402784] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Revised: 05/17/2024] [Accepted: 05/22/2024] [Indexed: 06/05/2024] Open
Abstract
Post-transcriptional regulation of immune-related transcripts by RNA-binding proteins (RBPs) impacts immune cell responses, including mast cell functionality. Despite their importance in immune regulation, the functional role of most RBPs remains to be understood. By manipulating the expression of specific RBPs in murine mast cells, coupled with mass spectrometry and transcriptomic analyses, we found that the Regnase family of proteins acts as a potent regulator of mast cell physiology. Specifically, Regnase-1 is required to maintain basic cell proliferation and survival, whereas both Regnase-1 and -3 cooperatively regulate the expression of inflammatory transcripts upon activation, with Tnf being a primary target in both human and mouse cells. Furthermore, Regnase-3 directly interacts with Regnase-1 in mast cells and is necessary to restrain Regnase-1 expression through the destabilization of its transcript. Overall, our study identifies protein interactors of endogenously expressed Regnase factors, characterizes the regulatory interplay between Regnase family members in mast cells, and establishes their role in the control of mast cell homeostasis and inflammatory responses.
Collapse
Affiliation(s)
- Marian Bataclan
- Institute for Research in Biomedicine, Università della Svizzera Italiana, Bellinzona, Switzerland
| | - Cristina Leoni
- Institute for Research in Biomedicine, Università della Svizzera Italiana, Bellinzona, Switzerland
| | - Simone G Moro
- Institute for Research in Biomedicine, Università della Svizzera Italiana, Bellinzona, Switzerland
| | - Matteo Pecoraro
- Institute for Research in Biomedicine, Università della Svizzera Italiana, Bellinzona, Switzerland
| | - Elaine H Wong
- Institute for Immunology, Biomedical Center, Faculty of Medicine, Ludwig-Maximilians-Universität in Munich, Planegg-Martinsried, Germany
| | - Vigo Heissmeyer
- Institute for Immunology, Biomedical Center, Faculty of Medicine, Ludwig-Maximilians-Universität in Munich, Planegg-Martinsried, Germany
- Research Unit Molecular Immune Regulation, Helmholtz Zentrum München, Munich, Germany
| | - Silvia Monticelli
- Institute for Research in Biomedicine, Università della Svizzera Italiana, Bellinzona, Switzerland
| |
Collapse
|
2
|
Franke K, Li Z, Bal G, Zuberbier T, Babina M. Synergism between IL-33 and MRGPRX2/FcεRI Is Primarily Due to the Complementation of Signaling Modules, and Only Modestly Supplemented by Prolonged Activation of Selected Kinases. Cells 2023; 12:2700. [PMID: 38067128 PMCID: PMC10705352 DOI: 10.3390/cells12232700] [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: 09/13/2023] [Revised: 11/16/2023] [Accepted: 11/19/2023] [Indexed: 12/18/2023] Open
Abstract
Skin mast cells (MCs) express high levels of MRGPRX2, FcεRI, and ST2, and vigorously respond to their ligands when triggered individually. IL-33/ST2 also potently synergizes with other receptors, but the molecular underpinnings are poorly understood. Human skin-derived MCs were stimulated via different receptors individually or jointly in the presence/absence of selective inhibitors. TNF was quantified by ELISA. Signaling cascades were studied by immunoblot. TNF was stimulated by FcεRI ≈ ST2 > MRGPRX2. Surprisingly, neither FcεRI nor MRGPRX2 stimulation elicited NF-κB activation (IκB degradation, p65 phosphorylation) in stark contrast to IL-33. Accordingly, TNF production did not depend on NF-κB in FcεRI- or MRGPRX2-stimulated MCs, but did well so downstream of ST2. Conversely, ERK1/2 and PI3K were the crucial modules upon FcεRI/MRGPRX2 stimulation, while p38 was key to the IL-33-elicited route. The different signaling prerequisites were mirrored by their activation patterns with potent pERK/pAKT after FcεRI/MRGPRX2, but preferential induction of pp38/NF-κB downstream of ST2. FcεRI/MRGPRX2 strongly synergized with IL-33, and some synergy was still observed upon inhibition of each module (ERK1/2, JNK, p38, PI3K, NF-κB). IL-33's contribution to synergism was owed to p38 > JNK > NF-κB, while the partner receptor contributed through ERK > PI3K ≈ JNK. Concurrent IL-33 led to slightly prolonged pERK (downstream of MRGPRX2) or pAKT (activated by FcεRI), while the IL-33-elicited modules (pp38/NF-κB) remained unaffected by co-stimulation of FcεRI/MRGPRX2. Collectively, the strong synergistic activity of IL-33 primarily results from the complementation of highly distinct modules following co-activation of the partner receptor rather than by altered signal strength of the same modules.
Collapse
Affiliation(s)
- Kristin Franke
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Immunology and Allergology IA, 12203 Berlin, Germany; (K.F.); (Z.L.); (G.B.); (T.Z.)
- Institute of Allergology, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt Universität zu Berlin, Hindenburgdamm 30, 12203 Berlin, Germany
| | - Zhuoran Li
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Immunology and Allergology IA, 12203 Berlin, Germany; (K.F.); (Z.L.); (G.B.); (T.Z.)
- Institute of Allergology, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt Universität zu Berlin, Hindenburgdamm 30, 12203 Berlin, Germany
| | - Gürkan Bal
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Immunology and Allergology IA, 12203 Berlin, Germany; (K.F.); (Z.L.); (G.B.); (T.Z.)
- Institute of Allergology, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt Universität zu Berlin, Hindenburgdamm 30, 12203 Berlin, Germany
| | - Torsten Zuberbier
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Immunology and Allergology IA, 12203 Berlin, Germany; (K.F.); (Z.L.); (G.B.); (T.Z.)
- Institute of Allergology, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt Universität zu Berlin, Hindenburgdamm 30, 12203 Berlin, Germany
| | - Magda Babina
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Immunology and Allergology IA, 12203 Berlin, Germany; (K.F.); (Z.L.); (G.B.); (T.Z.)
- Institute of Allergology, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt Universität zu Berlin, Hindenburgdamm 30, 12203 Berlin, Germany
| |
Collapse
|
3
|
Huang Y, Zhu Z, Li W, Ge Y, Li Y, Wang J, Peng X, Lin L, Li J, Liu CY, Li L. ELK4 exerts opposite roles in cytokine/chemokine production and degranulation in activated mast cells. Front Immunol 2023; 14:1171380. [PMID: 37529050 PMCID: PMC10389778 DOI: 10.3389/fimmu.2023.1171380] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Accepted: 06/14/2023] [Indexed: 08/03/2023] Open
Abstract
The proliferative potential of mast cells after activation for 3-4h was found to be decreased, which suggests that mast cell degranulation and cell proliferation are differentially regulated. ELK4, a member of the ternary complex factor (TCF) subfamily of Ets transcription factors, is one of the downstream effectors of MAPK signaling that is critical for cell proliferation. And Elk4 has been identified to be vital for macrophage activation in response to zymosan and the transcriptional response to 12-O-tetrade canoyl phorbol-13-acetate (TPA) stimulation in fibroblast. However, the effect of ELK4 on the mast cell transcriptional response to FcϵRI and GPCR mediated activation and its potential functional significance in mast cells remain unclear. Here, we showed that ELK4 expression is downregulated in activated mast cells. Elk4 knockout suppresses cell proliferation and impedes the cell cycle in bone marrow-derived mast cells (BMMCs), which is associated with decreased transcription of cell cycle genes. Additionally, the transcriptional activation of cytokines and chemokines is diminished while mast cell degranulation is enhanced in Elk4 knockout BMMCs. Mechanistically, ELK4 might positively modulate Hdc, Ccl3 and Ccl4 transcription by interacting with MITF and negatively regulate the transcription of degranulation-related genes by complexing with SIRT6. Overall, our study identifies a new physiological role of the transcription factor ELK4 in mast cell proliferation and activation.
Collapse
Affiliation(s)
- Yuji Huang
- Department of Laboratory Medicine, Shanghai General Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China
| | - Zhehui Zhu
- Department of Colorectal Surgery, Shanghai Engineering Research Center of Colorectal Cancer Minimally Invasive Technology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Weize Li
- Department of Laboratory Medicine, Shanghai General Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China
| | - Yiqin Ge
- Department of Laboratory Medicine, Shanghai General Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China
| | - Yanning Li
- Department of Laboratory Medicine, Shanghai General Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China
| | - Juan Wang
- Department of Laboratory Medicine, Shanghai General Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China
| | - Xia Peng
- Department of Laboratory Medicine, Shanghai General Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China
| | - Lihui Lin
- Department of Laboratory Medicine, Shanghai General Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China
| | - Jia Li
- Department of Laboratory Medicine, Shanghai General Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China
| | - Chen-Ying Liu
- Department of Colorectal and Anal Surgery, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Li Li
- Department of Laboratory Medicine, Shanghai General Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China
| |
Collapse
|
4
|
DeVore SB, Khurana Hershey GK. The role of the CBM complex in allergic inflammation and disease. J Allergy Clin Immunol 2022; 150:1011-1030. [PMID: 35981904 PMCID: PMC9643607 DOI: 10.1016/j.jaci.2022.06.023] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Revised: 06/15/2022] [Accepted: 06/30/2022] [Indexed: 10/15/2022]
Abstract
The caspase activation and recruitment domain-coiled-coil (CARD-CC) family of proteins-CARD9, CARD10, CARD11, and CARD14-is collectively expressed across nearly all tissues of the body and is a crucial mediator of immunologic signaling as part of the CARD-B-cell lymphoma/leukemia 10-mucosa-associated lymphoid tissue lymphoma translocation protein 1 (CBM) complex. Dysfunction or dysregulation of CBM proteins has been linked to numerous clinical manifestations known as "CBM-opathies." The CBM-opathy spectrum encompasses diseases ranging from mucocutaneous fungal infections and psoriasis to combined immunodeficiency and lymphoproliferative diseases; however, there is accumulating evidence that the CARD-CC family members also contribute to the pathogenesis and progression of allergic inflammation and allergic diseases. Here, we review the 4 CARD-CC paralogs, as well as B-cell lymphoma/leukemia 10 and mucosa-associated lymphoid tissue lymphoma translocation protein 1, and their individual and collective roles in the pathogenesis and progression of allergic inflammation and 4 major allergic diseases (allergic asthma, atopic dermatitis, food allergy, and allergic rhinitis).
Collapse
Affiliation(s)
- Stanley B DeVore
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio; Division of Asthma Research, Cincinnati Children's Hospital Medical Center, Cincinnati, Cincinnati, Ohio
| | - Gurjit K Khurana Hershey
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio; Division of Asthma Research, Cincinnati Children's Hospital Medical Center, Cincinnati, Cincinnati, Ohio.
| |
Collapse
|
5
|
Xue S, Rogers LR, Zheng M, He J, Piermarocchi C, Mias GI. Applying differential network analysis to longitudinal gene expression in response to perturbations. Front Genet 2022; 13:1026487. [PMID: 36324501 PMCID: PMC9618823 DOI: 10.3389/fgene.2022.1026487] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Accepted: 10/03/2022] [Indexed: 11/17/2022] Open
Abstract
Differential Network (DN) analysis is a method that has long been used to interpret changes in gene expression data and provide biological insights. The method identifies the rewiring of gene networks in response to external perturbations. Our study applies the DN method to the analysis of RNA-sequencing (RNA-seq) time series datasets. We focus on expression changes: (i) in saliva of a human subject after pneumococcal vaccination (PPSV23) and (ii) in primary B cells treated ex vivo with a monoclonal antibody drug (Rituximab). The DN method enabled us to identify the activation of biological pathways consistent with the mechanisms of action of the PPSV23 vaccine and target pathways of Rituximab. The community detection algorithm on the DN revealed clusters of genes characterized by collective temporal behavior. All saliva and some B cell DN communities showed characteristic time signatures, outlining a chronological order in pathway activation in response to the perturbation. Moreover, we identified early and delayed responses within network modules in the saliva dataset and three temporal patterns in the B cell data.
Collapse
Affiliation(s)
- Shuyue Xue
- Department of Physics and Astronomy, Michigan State University, East Lansing, MI, United States
- Institute for Quantitative Health Science and Engineering, Michigan State University, East Lansing, MI, United States
| | - Lavida R.K. Rogers
- Department of Biological Sciences, University of the Virgin Islands, St Thomas, US Virgin Islands
| | - Minzhang Zheng
- Institute for Quantitative Health Science and Engineering, Michigan State University, East Lansing, MI, United States
| | - Jin He
- Department of Biochemistry and Molecular Biology, Michigan State University, East Lansing, MI, United States
| | - Carlo Piermarocchi
- Department of Physics and Astronomy, Michigan State University, East Lansing, MI, United States
| | - George I. Mias
- Department of Physics and Astronomy, Michigan State University, East Lansing, MI, United States
- Institute for Quantitative Health Science and Engineering, Michigan State University, East Lansing, MI, United States
- Department of Biochemistry and Molecular Biology, Michigan State University, East Lansing, MI, United States
| |
Collapse
|
6
|
Ramakrishnan RK, Bajbouj K, Guimei M, Rawat SS, Kalaji Z, Hachim MY, Mahboub B, Ibrahim SM, Hamoudi R, Halwani R, Hamid Q. Bcl10 Regulates Lipopolysaccharide-Induced Pro-Fibrotic Signaling in Bronchial Fibroblasts from Severe Asthma Patients. Biomedicines 2022; 10:biomedicines10071716. [PMID: 35885021 PMCID: PMC9312497 DOI: 10.3390/biomedicines10071716] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 06/24/2022] [Accepted: 06/30/2022] [Indexed: 11/17/2022] Open
Abstract
Subepithelial fibrosis is a characteristic hallmark of airway remodeling in asthma. Current asthma medications have limited efficacy in treating fibrosis, particularly in patients with severe asthma, necessitating a deeper understanding of the fibrotic mechanisms. The NF-κB pathway is key to airway inflammation in asthma, as it regulates the activity of multiple pro-inflammatory mediators that contribute to airway pathology. Bcl10 is a well-known upstream mediator of the NF-κB pathway that has been linked to fibrosis in other disease models. Therefore, we investigated Bcl10-mediated NF-κB activation as a potential pathway regulating fibrotic signaling in severe asthmatic fibroblasts. We demonstrate here the elevated protein expression of Bcl10 in bronchial fibroblasts and bronchial biopsies from severe asthmatic patients when compared to non-asthmatic individuals. Lipopolysaccharide (LPS) induced the increased expression of the pro-fibrotic cytokines IL-6, IL-8 and TGF-β1 in bronchial fibroblasts, and this induction was associated with the activation of Bcl10. Inhibition of the Bcl10-mediated NF-κB pathway using an IRAK1/4 selective inhibitor abrogated the pro-fibrotic signaling induced by LPS. Thus, our study indicates that Bcl10-mediated NF-κB activation signals increased pro-fibrotic cytokine expression in severe asthmatic airways. This reveals the therapeutic potential of targeting Bcl10 signaling in ameliorating inflammation and fibrosis, particularly in severe asthmatic individuals.
Collapse
Affiliation(s)
- Rakhee K. Ramakrishnan
- Sharjah Institute for Medical Research, College of Medicine, University of Sharjah, Sharjah P.O. Box 27272, United Arab Emirates; (R.K.R.); (K.B.); (Z.K.); (B.M.); (S.M.I.)
| | - Khuloud Bajbouj
- Sharjah Institute for Medical Research, College of Medicine, University of Sharjah, Sharjah P.O. Box 27272, United Arab Emirates; (R.K.R.); (K.B.); (Z.K.); (B.M.); (S.M.I.)
| | - Maha Guimei
- Department of Pathology, Faculty of Medicine, Alexandria University, Alexandria 21526, Egypt;
| | - Surendra Singh Rawat
- College of Medicine, Mohammed Bin Rashid University, Dubai P.O. Box 505055, United Arab Emirates; (S.S.R.); (M.Y.H.)
| | - Zaina Kalaji
- Sharjah Institute for Medical Research, College of Medicine, University of Sharjah, Sharjah P.O. Box 27272, United Arab Emirates; (R.K.R.); (K.B.); (Z.K.); (B.M.); (S.M.I.)
| | - Mahmood Y. Hachim
- College of Medicine, Mohammed Bin Rashid University, Dubai P.O. Box 505055, United Arab Emirates; (S.S.R.); (M.Y.H.)
| | - Bassam Mahboub
- Sharjah Institute for Medical Research, College of Medicine, University of Sharjah, Sharjah P.O. Box 27272, United Arab Emirates; (R.K.R.); (K.B.); (Z.K.); (B.M.); (S.M.I.)
- Rashid Hospital, Dubai Health Authority, Dubai P.O. Box 4545, United Arab Emirates
| | - Saleh M. Ibrahim
- Sharjah Institute for Medical Research, College of Medicine, University of Sharjah, Sharjah P.O. Box 27272, United Arab Emirates; (R.K.R.); (K.B.); (Z.K.); (B.M.); (S.M.I.)
- Lübeck Institute of Experimental Dermatology (LIED), University of Lübeck, 23562 Lübeck, Germany
| | - Rifat Hamoudi
- Sharjah Institute for Medical Research, College of Medicine, University of Sharjah, Sharjah P.O. Box 27272, United Arab Emirates; (R.K.R.); (K.B.); (Z.K.); (B.M.); (S.M.I.)
- Division of Surgery and Interventional Science, University College London, London WC1E 6BT, UK
- Correspondence: (R.H.); (R.H.); (Q.H.)
| | - Rabih Halwani
- Sharjah Institute for Medical Research, College of Medicine, University of Sharjah, Sharjah P.O. Box 27272, United Arab Emirates; (R.K.R.); (K.B.); (Z.K.); (B.M.); (S.M.I.)
- Immunology Research Lab, College of Medicine, King Saud University, Riyadh P.O. Box 145111, Saudi Arabia
- Correspondence: (R.H.); (R.H.); (Q.H.)
| | - Qutayba Hamid
- Sharjah Institute for Medical Research, College of Medicine, University of Sharjah, Sharjah P.O. Box 27272, United Arab Emirates; (R.K.R.); (K.B.); (Z.K.); (B.M.); (S.M.I.)
- Meakins-Christie Laboratories, McGill University, Montreal, QC H3A 0G4, Canada
- Correspondence: (R.H.); (R.H.); (Q.H.)
| |
Collapse
|
7
|
Zhang YY, Peng J, Luo XJ. Post-translational modification of MALT1 and its role in B cell- and T cell-related diseases. Biochem Pharmacol 2022; 198:114977. [PMID: 35218741 DOI: 10.1016/j.bcp.2022.114977] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2022] [Revised: 02/18/2022] [Accepted: 02/18/2022] [Indexed: 02/06/2023]
Abstract
Mucosa-associated lymphoid tissue lymphoma translocation protein 1 (MALT1) is a multifunctional protein. MALT1 functions as an adaptor protein to assemble and recruit proteins such as B-cell lymphoma 10 (BCL10) and caspase-recruitment domain (CARD)-containing coiled-coil protein 11 (CARD11). Conversely it also acts as a paracaspase to cleave specified substrates. Because of its involvement in immunity, inflammation and cancer through its dual functions of scaffolding and catalytic activity, MALT1 is becoming a promising therapeutic target in B cell- and T cell-related diseases. There is growing evidence that the function of MALT1 is subtly modulated via post-translational modifications. This review summarized recent progress in relevant studies regarding the physiological and pathophysiological functions of MALT1, post-translational modifications of MALT1 and its role in B cell- and T cell- related diseases. In addition, the current available MALT1 inhibitors were also discussed.
Collapse
Affiliation(s)
- Yi-Yue Zhang
- Department of Pharmacology, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410078, China
| | - Jun Peng
- Department of Pharmacology, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410078, China; Hunan Provincial Key Laboratory of Cardiovascular Research, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410078, China.
| | - Xiu-Ju Luo
- Department of Laboratory Medicine, The Third Xiangya Hospital of Central South University, Changsha 410013, China.
| |
Collapse
|
8
|
Li R, Chen S, Gu X, An S, Wang Z. Role of the nuclear receptor subfamily 4a in mast cells in the development of irritable bowel syndrome. Comput Struct Biotechnol J 2022; 20:1198-1207. [PMID: 35317226 PMCID: PMC8907967 DOI: 10.1016/j.csbj.2022.02.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Revised: 02/17/2022] [Accepted: 02/19/2022] [Indexed: 11/15/2022] Open
Abstract
The activation of mast cells (MCs) and mediator release are closely related to the pathophysiology of irritable bowel syndrome (IBS). However, the exact underlying mechanisms are still not completely understood. The nuclear receptor subfamily 4a (Nr4a) is a family of orphan nuclear receptors implicated in regulating MC activation, degranulation, cytokine/chemokine synthesis and release. Acute and chronic stress trigger hypothalamic–pituitaryadrenal axis (HPA) activation to induce the release of corticotropin-releasing hormone (CRH), resulting in MC activation and induction of the Nr4a family. Our newest data showed that Nr4a members were specially over-expressed in colonic MCs of the chronic water-avoidance stress (WAS)-induced visceral hyperalgesia mice, suggesting that Nr4a members might be involved in the pathophysiology of visceral hypersensitivity. In this review, we highlight the present knowledge on roles of Nr4a members in the activation of MCs and the pathophysiology of IBS, and discuss signaling pathways that modulate the activation of Nr4a family members. We propose that a better understanding of Nr4a members and their modulators may facilitate the development of more selective and effective therapies to treat IBS patients.
Collapse
Affiliation(s)
| | | | | | - Shuhong An
- Corresponding authors at: Department of Human Anatomy, Shandong First Medical University & Shandong Academy of Medical Sciences, 2 Ying Sheng Dong Lu, Taian 271000, China.
| | - Zhaojin Wang
- Corresponding authors at: Department of Human Anatomy, Shandong First Medical University & Shandong Academy of Medical Sciences, 2 Ying Sheng Dong Lu, Taian 271000, China.
| |
Collapse
|
9
|
Wang Y, Yi K, Liu X, Tan Y, Jin W, Li Y, Zhou J, Wang H, Kang C. HOTAIR Up-Regulation Activates NF-κB to Induce Immunoescape in Gliomas. Front Immunol 2021; 12:785463. [PMID: 34887871 PMCID: PMC8649724 DOI: 10.3389/fimmu.2021.785463] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Accepted: 11/05/2021] [Indexed: 12/20/2022] Open
Abstract
Background Checkpoint blockade therapies targeting programmed death ligand 1 (PD-L1) and its receptor programmed cell death 1 promote T cell-mediated immune surveillance against tumors and have been associated with significant clinical benefit in cancer patients. The long-stranded non-coding RNA HOTAIR is highly expressed and associated with metastasis in a variety of cancer types and promotes tumor metastasis at least in part through association with the PRC2 complex that induces redirection to hundreds of genes involved in tumor metastasis. Here, we report that HOTAIR is an activator lncRNA of the NF-κB pathway and demonstrate that its apparent upregulation promotes inflammatory signaling and immune escape in glioma cells. Methods Bioinformatics analysis was used to elucidate the relationship between HOTAIR and NF-κB pathway in HOTAIR knockdown glioma cells. At the cytological level, protein hybridization and immunofluorescence were used to detect the response of proteins in the NF-κB signaling pathway to HOTAIR regulation. ChIP and ChIRP experiments identified HOTAIR target genes. Animal experiments verified alterations in inflammation and immune escape following HOTAIR knockdown and activity inhibition. Results HOTAIR activated the expression of proteins involved in NF-κB, TNFα, MAPK and other inflammatory signaling pathways. In addition, HOTAIR induced various proteins containing protein kinase structural domains and promoted the enrichment of proteins and complexes of important inflammatory signaling pathways, such as the TNFα/NF-κB signaling protein complex, the IκB kinase complex, and the IKKA-IKKB complex. In addition, HOTAIR aberrantly activated biological processes involved in glioma immune responses, T-cell co-stimulation and transcription initiation by RNA polymerase II. HOTAIR facilitated the induction of IκBα phosphorylation by suppressing the expression of the NF-κB upstream protein UBXN1, promoting NF-κB phosphorylation and nuclear translocation. In vivo, reduction of HOTAIR decreased PD-L1 protein expression, indicating that cells are more likely to be targeted by immune T cells. Conclusion In conclusion, our results provide convincing evidence that lncRNA HOTAIR drives aberrant gene transcription and immune escape from tumor cells through the NF-κB pathway.
Collapse
Affiliation(s)
- Yunfei Wang
- Laboratory of Neuro-Oncology, Department of Neurosurgery, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, China.,Key Laboratory of Post-Neuro Injury Neuro-Repair and Regeneration in Central Nervous System, Ministry of Education and Tianjin City, Tianjin, China
| | - Kaikai Yi
- Laboratory of Neuro-Oncology, Department of Neurosurgery, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, China.,Key Laboratory of Post-Neuro Injury Neuro-Repair and Regeneration in Central Nervous System, Ministry of Education and Tianjin City, Tianjin, China.,Department of Neuro-Oncology and Neurosurgery, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China.,National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy of Tianjin, Tianjin Clinical Research Center for Cancer, Tianjin, China
| | - Xing Liu
- Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
| | - Yanli Tan
- Department of Pathology, Hebei University Medical College, Baoding, China.,Department of Pathology, Affiliated Hospital of Hebei University, Baoding, China
| | - Weili Jin
- Laboratory of Neuro-Oncology, Department of Neurosurgery, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, China.,Key Laboratory of Post-Neuro Injury Neuro-Repair and Regeneration in Central Nervous System, Ministry of Education and Tianjin City, Tianjin, China
| | - Yansheng Li
- Laboratory of Neuro-Oncology, Department of Neurosurgery, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, China.,Key Laboratory of Post-Neuro Injury Neuro-Repair and Regeneration in Central Nervous System, Ministry of Education and Tianjin City, Tianjin, China
| | - Junhu Zhou
- Laboratory of Neuro-Oncology, Department of Neurosurgery, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, China.,Key Laboratory of Post-Neuro Injury Neuro-Repair and Regeneration in Central Nervous System, Ministry of Education and Tianjin City, Tianjin, China
| | - Hongjun Wang
- Department of Neurosurgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Chunsheng Kang
- Laboratory of Neuro-Oncology, Department of Neurosurgery, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, China.,Key Laboratory of Post-Neuro Injury Neuro-Repair and Regeneration in Central Nervous System, Ministry of Education and Tianjin City, Tianjin, China
| |
Collapse
|
10
|
Leong E, Pang Z, Stadnyk AW, Lin TJ. Calcineurin Aα Contributes to IgE-Dependent Mast-Cell Mediator Secretion in Allergic Inflammation. J Innate Immun 2021; 14:320-334. [PMID: 34839285 PMCID: PMC9274814 DOI: 10.1159/000520040] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2019] [Accepted: 09/26/2021] [Indexed: 11/19/2022] Open
Abstract
Mast cells (MCs) are key mediators of allergic inflammation through the activation of cross-linked immunoglobulin E (IgE) bound to the high-affinity IgE receptor (FcϵRI) on the cell surface, leading to the release of biologically potent mediators, either from preformed granules or newly synthesized. Pharmacological inhibitors have been developed to target a key signaling protein phosphatase in this pathway, calcineurin, yet there is a lack of genetic and definitive evidence for the various isoforms of calcineurin subunits in FcϵRI-mediated responses. In this study, we hypothesized that deficiency in the calcineurin Aα isoform will result in a decreased allergic immune response by the MCs. In a model of passive cutaneous anaphylaxis, there was a reduction in vascular permeability in MC-deficient mouse tissues reconstituted with calcineurin subunit A (CnAα) gene-knockout (CnAα<sup>−/−</sup>) MCs, and in vitro experiments identified a significant reduction in release of preformed mediators from granules. Furthermore, released levels of de novo synthesized cytokines were reduced upon FcϵRI activation of CnAα<sup>−/−</sup> MCs in vitro. Characterizing the mechanisms associated with this deficit response, we found a significant impairment of nuclear factor of kappa light polypeptide gene enhancer in B cell phosphorylation and impaired nuclear factor kappa-light-chain-enhancer of activated B-cell inhibitor alpha (NF-κB) activation. Thus, we concluded that CnAα contributes to the release of preformed mediators and newly synthesized mediators from FcϵRI-mediated activation of MCs, and this regulation includes NF-κB signaling.
Collapse
Affiliation(s)
- Edwin Leong
- Department of Pathology, Dalhousie University, Halifax, Nova Scotia, Canada,
| | - Zheng Pang
- Department of Pathology, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Andrew W Stadnyk
- Department of Pathology, Dalhousie University, Halifax, Nova Scotia, Canada.,Department of Pediatrics, Isaac Walton Killam Health Centre, Halifax, Nova Scotia, Canada.,Department of Microbiology & Immunology, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Tong-Jun Lin
- Department of Pathology, Dalhousie University, Halifax, Nova Scotia, Canada.,Department of Pediatrics, Isaac Walton Killam Health Centre, Halifax, Nova Scotia, Canada.,Department of Microbiology & Immunology, Dalhousie University, Halifax, Nova Scotia, Canada
| |
Collapse
|
11
|
Bataclan M, Leoni C, Monticelli S. RNA-binding proteins and RNA methylation in myeloid cells. Immunol Rev 2021; 304:51-61. [PMID: 34523134 PMCID: PMC7615035 DOI: 10.1111/imr.13025] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Revised: 08/26/2021] [Accepted: 09/02/2021] [Indexed: 11/30/2022]
Abstract
RNA-binding proteins (RBPs) regulate all aspects of the life of mRNA transcripts. They are critically important in regulating immune responses, most notably by restraining excessive inflammation that can potentially lead to tissue damage. RBPs are also crucial for pathogen sensing, for instance for the recognition of viral nucleic acids. Concordant with these central regulatory roles, the dysregulated activity of many RBPs can give rise to disease. The expression and function of RBPs are therefore highly controlled by an elaborate network of transcriptional, post-transcriptional and post-translational mechanisms, including the ability of different RBPs to cross-regulate each other's expression. With an emphasis on macrophages and mast cells, we review current knowledge on the role of selected RBPs that have been shown to directly impact the expression of inflammatory transcripts. By focusing specifically on proteins of the Regnase and ZFP36 family, as well as on factors involved in N6 -methyladenosine (m6 A) deposition and recognition, we discuss mechanism of action, regulatory feedback, and impact of these selected proteins on immune responses. Finally, we include examples of the role of m6 A and RBPs in the recognition of viral RNAs. Overall, we provide a general overview of the impact of selected RBPs on the myeloid compartment, followed by a discussion of outstanding questions and challenges for the future.
Collapse
Affiliation(s)
- Marian Bataclan
- Institute for Research in Biomedicine, Università della Svizzera italiana, Via Vincenzo Vela 6, CH-6500 Bellinzona, Switzerland
| | - Cristina Leoni
- Institute for Research in Biomedicine, Università della Svizzera italiana, Via Vincenzo Vela 6, CH-6500 Bellinzona, Switzerland
| | - Silvia Monticelli
- Institute for Research in Biomedicine, Università della Svizzera italiana, Via Vincenzo Vela 6, CH-6500 Bellinzona, Switzerland
| |
Collapse
|
12
|
Zhang Z, Kurashima Y. Two Sides of the Coin: Mast Cells as a Key Regulator of Allergy and Acute/Chronic Inflammation. Cells 2021; 10:cells10071615. [PMID: 34203383 PMCID: PMC8308013 DOI: 10.3390/cells10071615] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Revised: 06/18/2021] [Accepted: 06/25/2021] [Indexed: 12/17/2022] Open
Abstract
It is well known that mast cells (MCs) initiate type I allergic reactions and inflammation in a quick response to the various stimulants, including—but not limited to—allergens, pathogen-associated molecular patterns (PAMPs), and damage-associated molecular patterns (DAMPs). MCs highly express receptors of these ligands and proteases (e.g., tryptase, chymase) and cytokines (TNF), and other granular components (e.g., histamine and serotonin) and aggravate the allergic reaction and inflammation. On the other hand, accumulated evidence has revealed that MCs also possess immune-regulatory functions, suppressing chronic inflammation and allergic reactions on some occasions. IL-2 and IL-10 released from MCs inhibit excessive immune responses. Recently, it has been revealed that allergen immunotherapy modulates the function of MCs from their allergic function to their regulatory function to suppress allergic reactions. This evidence suggests the possibility that manipulation of MCs functions will result in a novel approach to the treatment of various MCs-mediated diseases.
Collapse
Affiliation(s)
- Zhongwei Zhang
- Department of Innovative Medicine, Graduate School of Medicine, Chiba University, Chiba 260-8670, Japan;
| | - Yosuke Kurashima
- Department of Innovative Medicine, Graduate School of Medicine, Chiba University, Chiba 260-8670, Japan;
- Department of Mucosal Immunology, The University of Tokyo Distinguished Professor Unit, The Institute of Medical Science, The University of Tokyo, Tokyo 108-8639, Japan
- International Research and Development Center for Mucosal Vaccines, The Institute of Medical Science, The University of Tokyo, Tokyo 108-8639, Japan
- CU-UCSD Center for Mucosal Immunology, Department of Pathology/Medicine, Allergy and Vaccines, University of California, San Diego, CA 92093-0063, USA
- Mucosal Immunology and Allergy Therapeutics, Institute for Global Prominent Research, Graduate School of Medicine, Chiba University, Chiba 260-8670, Japan
- Correspondence: ; Tel.: +81-43-226-2848; Fax: +81-43-226-2183
| |
Collapse
|
13
|
Natarajan S, Govender K, Shobo A, Baijnath S, Arvidsson PI, Govender T, Lin J, Maguire GE, Naicker T, Kruger HG. Potential of brain mast cells for therapeutic application in the immune response to bacterial and viral infections. Brain Res 2021; 1767:147524. [PMID: 34015358 DOI: 10.1016/j.brainres.2021.147524] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Revised: 04/21/2021] [Accepted: 05/11/2021] [Indexed: 10/21/2022]
Abstract
A wide range of microorganisms can infect the central nervous system (CNS). The immune response of the CNS provides limited protection against microbes penetrating the blood-brain barrier. This results in a neurological deficit and sometimes leads to high morbidity and mortality rates despite advanced therapies. For the last two decades, different studies have expanded our understanding of the molecular basis of human neuroinfectious diseases, especially concerning the contributions of mast cell interactions with other central nervous system compartments. Brain mast cells are multifunctional cells derived from the bone marrow and reside in the brain. Their proximity to blood vessels, their role as "first responders" their unique receptors systems and their ability to rapidly release pathogen responsive mediators enable them to exert a crucial defensive role in the host-defense system. This review describes key biological and physiological functions of mast cells, concerning their ability to recognize pathogens via various receptor systems, followed by a coordinated and selective mediator release upon specific interactions with pathogenic stimulating factors. The goal of this review is to direct attention to the possibilities for therapeutic applications of mast cells against bacterial and viral related infections. We also focus on opportunities for future research activating mast cells via adjuvants.
Collapse
Affiliation(s)
- Satheesh Natarajan
- Catalysis and Peptide Research Unit, University of KwaZulu-Natal, Westville Campus, E-Block, 6th Floor, Room E1-06-016, Durban, South Africa
| | - Kamini Govender
- Catalysis and Peptide Research Unit, University of KwaZulu-Natal, Westville Campus, E-Block, 6th Floor, Room E1-06-016, Durban, South Africa
| | - Adeola Shobo
- Catalysis and Peptide Research Unit, University of KwaZulu-Natal, Westville Campus, E-Block, 6th Floor, Room E1-06-016, Durban, South Africa
| | - Sooraj Baijnath
- Catalysis and Peptide Research Unit, University of KwaZulu-Natal, Westville Campus, E-Block, 6th Floor, Room E1-06-016, Durban, South Africa
| | - Per I Arvidsson
- Catalysis and Peptide Research Unit, University of KwaZulu-Natal, Westville Campus, E-Block, 6th Floor, Room E1-06-016, Durban, South Africa; Science for Life Laboratory, Drug Discovery and Development, Platform and Division of Translational Medicine and Chemical Biology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Sweden
| | - Thavendran Govender
- Department of Chemistry, University of Zululand, Private Bag X1001, KwaDlangezwa 3886, South Africa
| | - Johnson Lin
- School of Life Sciences, University of KwaZulu-Natal, Durban 4001, South Africa; School of Chemistry and Physics, University of KwaZulu-Natal, Durban 4001, South Africa
| | - Glenn Em Maguire
- Catalysis and Peptide Research Unit, University of KwaZulu-Natal, Westville Campus, E-Block, 6th Floor, Room E1-06-016, Durban, South Africa
| | - Tricia Naicker
- Catalysis and Peptide Research Unit, University of KwaZulu-Natal, Westville Campus, E-Block, 6th Floor, Room E1-06-016, Durban, South Africa
| | - Hendrik G Kruger
- Catalysis and Peptide Research Unit, University of KwaZulu-Natal, Westville Campus, E-Block, 6th Floor, Room E1-06-016, Durban, South Africa.
| |
Collapse
|
14
|
Zhang S, Edwards TN, Chaudhri VK, Wu J, Cohen JA, Hirai T, Rittenhouse N, Schmitz EG, Zhou PY, McNeil BD, Yang Y, Koerber HR, Sumpter TL, Poholek AC, Davis BM, Albers KM, Singh H, Kaplan DH. Nonpeptidergic neurons suppress mast cells via glutamate to maintain skin homeostasis. Cell 2021; 184:2151-2166.e16. [PMID: 33765440 PMCID: PMC8052305 DOI: 10.1016/j.cell.2021.03.002] [Citation(s) in RCA: 102] [Impact Index Per Article: 25.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Revised: 01/21/2021] [Accepted: 03/01/2021] [Indexed: 01/15/2023]
Abstract
Cutaneous mast cells mediate numerous skin inflammatory processes and have anatomical and functional associations with sensory afferent neurons. We reveal that epidermal nerve endings from a subset of sensory nonpeptidergic neurons expressing MrgprD are reduced by the absence of Langerhans cells. Loss of epidermal innervation or ablation of MrgprD-expressing neurons increased expression of a mast cell gene module, including the activating receptor, Mrgprb2, resulting in increased mast cell degranulation and cutaneous inflammation in multiple disease models. Agonism of MrgprD-expressing neurons reduced expression of module genes and suppressed mast cell responses. MrgprD-expressing neurons released glutamate which was increased by MrgprD agonism. Inhibiting glutamate release or glutamate receptor binding yielded hyperresponsive mast cells with a genomic state similar to that in mice lacking MrgprD-expressing neurons. These data demonstrate that MrgprD-expressing neurons suppress mast cell hyperresponsiveness and skin inflammation via glutamate release, thereby revealing an unexpected neuroimmune mechanism maintaining cutaneous immune homeostasis.
Collapse
Affiliation(s)
- Shiqun Zhang
- Department of Dermatology, University of Pittsburgh, Pittsburgh, PA 15261, USA; Department of Immunology, University of Pittsburgh, Pittsburgh, PA 15261, USA; Department of Neurobiology, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Tara N Edwards
- Department of Dermatology, University of Pittsburgh, Pittsburgh, PA 15261, USA; Department of Immunology, University of Pittsburgh, Pittsburgh, PA 15261, USA; Department of Neurobiology, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Virendra K Chaudhri
- Department of Immunology, University of Pittsburgh, Pittsburgh, PA 15261, USA; Center for Systems Immunology, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Jianing Wu
- Department of Dermatology, University of Pittsburgh, Pittsburgh, PA 15261, USA; Department of Immunology, University of Pittsburgh, Pittsburgh, PA 15261, USA; School of Medicine, Tsinghua University, No. 1 Tsinghua Yuan, Haidian District, Beijing 100084, China
| | - Jonathan A Cohen
- Department of Dermatology, University of Pittsburgh, Pittsburgh, PA 15261, USA; Department of Immunology, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Toshiro Hirai
- Department of Dermatology, University of Pittsburgh, Pittsburgh, PA 15261, USA; Department of Immunology, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Natalie Rittenhouse
- Division of Rheumatology, Department of Pediatrics, University of Pittsburgh, Pittsburgh, PA 15224, USA
| | - Elizabeth G Schmitz
- Division of Rheumatology, Department of Pediatrics, University of Pittsburgh, Pittsburgh, PA 15224, USA
| | - Paul Yifan Zhou
- Department of Dermatology, University of Pittsburgh, Pittsburgh, PA 15261, USA; Department of Immunology, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Benjamin D McNeil
- Division of Allergy & Immunology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Yi Yang
- Department of Dermatology, University of Pittsburgh, Pittsburgh, PA 15261, USA; Department of Immunology, University of Pittsburgh, Pittsburgh, PA 15261, USA; Department of Nuclear Medicine, Xiangya Hospital, Central South University, Changsha, Hunan 410013, China
| | - H Richard Koerber
- Pittsburgh Center for Pain Research, University of Pittsburgh, Pittsburgh, PA 15261, USA; Department of Neurobiology, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Tina L Sumpter
- Department of Dermatology, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Amanda C Poholek
- Department of Immunology, University of Pittsburgh, Pittsburgh, PA 15261, USA; Division of Rheumatology, Department of Pediatrics, University of Pittsburgh, Pittsburgh, PA 15224, USA
| | - Brian M Davis
- Pittsburgh Center for Pain Research, University of Pittsburgh, Pittsburgh, PA 15261, USA; Department of Neurobiology, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Kathryn M Albers
- Pittsburgh Center for Pain Research, University of Pittsburgh, Pittsburgh, PA 15261, USA; Department of Neurobiology, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Harinder Singh
- Department of Immunology, University of Pittsburgh, Pittsburgh, PA 15261, USA; Center for Systems Immunology, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Daniel H Kaplan
- Department of Dermatology, University of Pittsburgh, Pittsburgh, PA 15261, USA; Department of Immunology, University of Pittsburgh, Pittsburgh, PA 15261, USA; Pittsburgh Center for Pain Research, University of Pittsburgh, Pittsburgh, PA 15261, USA.
| |
Collapse
|
15
|
Yiu WH, Chan KW, Chan LYY, Leung JCK, Lai KN, Tang SCW. Spleen Tyrosine Kinase Inhibition Ameliorates Tubular Inflammation in IgA Nephropathy. Front Physiol 2021; 12:650888. [PMID: 33790807 PMCID: PMC8006276 DOI: 10.3389/fphys.2021.650888] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Accepted: 02/01/2021] [Indexed: 12/15/2022] Open
Abstract
Spleen tyrosine kinase (Syk) is a non-receptor tyrosine kinase involved in signal transduction in a variety of immune responses. It has been demonstrated that Syk plays a pathogenic role in orchestrating inflammatory responses and cell proliferation in human mesangial cells (HMC) in IgA nephropathy (IgAN). However, whether Syk is involved in tubular damage in IgAN remains unknown. Using human kidney biopsy specimens, we found that Syk was activated in renal tubules of biopsy-proven IgAN patients with an increase in total and phosphorylated levels compared to that from healthy control subjects. In vitro, cultured proximal tubular epithelial cells (PTECs) were stimulated with conditioned medium prepared from human mesangial cells incubated with polymeric IgA (IgA-HMC) from patients with IgAN or healthy control. Induction of IL-6, IL-8, and ICAM-1 synthesis from cultured PTECs incubated with IgA-HMC conditioned medium was significantly suppressed by treatment with the Syk inhibitor R406 compared to that from healthy control. Furthermore, R406 downregulated expression of phosphorylated p65 NF-κB and p-42/p-44 MAPK, and attenuated TNF-α-induced cytokine production in PTECs. Taken together, our findings suggest that Syk mediates IgA-HMC conditioned medium-induced inflammation in tubular cells via activation of NF-κB and p-42/p-44 MAPK signaling. Inhibition of Syk may be a potential therapeutic approach for tubulointerstitial injury in IgAN.
Collapse
Affiliation(s)
- Wai Han Yiu
- Department of Medicine, The University of Hong Kong, Queen Mary Hospital, Pokfulam, Hong Kong
| | - Kam Wa Chan
- Department of Medicine, The University of Hong Kong, Queen Mary Hospital, Pokfulam, Hong Kong
| | - Loretta Y Y Chan
- Department of Medicine, The University of Hong Kong, Queen Mary Hospital, Pokfulam, Hong Kong
| | - Joseph C K Leung
- Department of Medicine, The University of Hong Kong, Queen Mary Hospital, Pokfulam, Hong Kong
| | - Kar Neng Lai
- Department of Medicine, The University of Hong Kong, Queen Mary Hospital, Pokfulam, Hong Kong
| | - Sydney C W Tang
- Department of Medicine, The University of Hong Kong, Queen Mary Hospital, Pokfulam, Hong Kong
| |
Collapse
|
16
|
Han EJ, Kim HS, Sanjeewa KKA, Jung K, Jee Y, Jeon YJ, Fernando IPS, Ahn G. Sargassum horneri as a Functional Food Ameliorated IgE/BSA-Induced Mast Cell Activation and Passive Cutaneous Anaphylaxis in Mice. Mar Drugs 2020; 18:E594. [PMID: 33256200 PMCID: PMC7760094 DOI: 10.3390/md18120594] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Revised: 11/23/2020] [Accepted: 11/25/2020] [Indexed: 12/21/2022] Open
Abstract
Sargassum horneri (S. horneri), an edible brown alga, has been proposed as a functional food with an improvement effect on abnormal skin immune responses. The present study investigates the anti-allergic effect of an ethanol extract from S. horneri (SHE) on immunoglobulin E (IgE)/bovine serum albumin (BSA)-mediated activation in bone marrow-derived cultured-mast cells (BMCMCs) and passive cutaneous anaphylaxis (PCA) reaction in mice. SHE markedly and dose-dependently suppressed the degranulation of BMCMCs by reducing the β-hexosaminidase and histamine release without cytotoxicity. In addition, SHE significantly decreased the FcεRI expression on the surface of BMCMCs and its IgE binding. Moreover, SHE reduced the mRNA expression and the production of allergic cytokines; interleukin (IL)-1β, IL-4, IL-5, IL-6, IL-10, IL-13; interferon (IFN)-γ and/or tumor necrosis factor (TNF)-α; and a chemokine, thymus and activation-regulated chemokine (TARC), by suppressing the activation of Src-family kinases and nuclear factor (NF)-κB signaling. In further study, the application of SHE reduced the PCA reaction in an IgE/BSA-induced type I allergic mice model. Taken together, we suggest that SHE has an anti-allergic effect in type I allergic responses.
Collapse
Affiliation(s)
- Eui Jeong Han
- Research Center for Healthcare and Biomedical Engineering, Chonnam National University, Yeosu 59626, Korea;
- Department of Food Technology and Nutrition, Chonnam National University, Yeosu 59626, Korea
| | - Hyun-Soo Kim
- National Marine Biodiversity Institute of Korea, Janghang-eup, Seocheon 33662, Korea;
| | - Kalu Kapuge Asanka Sanjeewa
- Department of Marine Life Science, School of Marine Biomedical Sciences, Jeju National University, Jeju 63243, Korea; (K.K.A.S.); (Y.-J.J.)
| | - Kyungsook Jung
- Biomaterials Research Center, Korea Research Institute of Bioscience and Biotechnology, Jeonbuk 56212, Korea;
| | - Youngheun Jee
- Department of Veterinary Medicine and Veterinary Medical Research Institute, Jeju National University, Jeju 63243, Korea;
- Interdisciplinary Graduate Program in Advanced Convergence Technology & Science, Jeju National University, Jeju 63243, Korea
| | - You-Jin Jeon
- Department of Marine Life Science, School of Marine Biomedical Sciences, Jeju National University, Jeju 63243, Korea; (K.K.A.S.); (Y.-J.J.)
| | - Ilekuttige Priyan Shanura Fernando
- Department of Marine Bio-Food Sciences, Chonnam National University, Yeosu 59626, Korea;
- Control Center for Aquatic Animal Diseases, Chonnam National University, Yeosu 59626, Korea
| | - Ginnae Ahn
- Department of Food Technology and Nutrition, Chonnam National University, Yeosu 59626, Korea
- Department of Marine Bio-Food Sciences, Chonnam National University, Yeosu 59626, Korea;
| |
Collapse
|
17
|
The Utility of Resolving Asthma Molecular Signatures Using Tissue-Specific Transcriptome Data. G3-GENES GENOMES GENETICS 2020; 10:4049-4062. [PMID: 32900903 PMCID: PMC7642926 DOI: 10.1534/g3.120.401718] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
An integrative analysis focused on multi-tissue transcriptomics has not been done for asthma. Tissue-specific DEGs remain undetected in many multi-tissue analyses, which influences identification of disease-relevant pathways and potential drug candidates. Transcriptome data from 609 cases and 196 controls, generated using airway epithelium, bronchial, nasal, airway macrophages, distal lung fibroblasts, proximal lung fibroblasts, CD4+ lymphocytes, CD8+ lymphocytes from whole blood and induced sputum samples, were retrieved from Gene Expression Omnibus (GEO). Differentially regulated asthma-relevant genes identified from each sample type were used to identify (a) tissue-specific and tissue-shared asthma pathways, (b) their connection to GWAS-identified disease genes to identify candidate tissue for functional studies, (c) to select surrogate sample for invasive tissues, and finally (d) to identify potential drug candidates via connectivity map analysis. We found that inter-tissue similarity in gene expression was more pronounced at pathway/functional level than at gene level with highest similarity between bronchial epithelial cells and lung fibroblasts, and lowest between airway epithelium and whole blood samples. Although public-domain gene expression data are limited by inadequately annotated per-sample demographic and clinical information which limited the analysis, our tissue-resolved analysis clearly demonstrated relative importance of unique and shared asthma pathways, At the pathway level, IL-1b signaling and ERK signaling were significant in many tissue types, while Insulin-like growth factor and TGF-beta signaling were relevant in only airway epithelial tissue. IL-12 (in macrophages) and Immunoglobulin signaling (in lymphocytes) and chemokines (in nasal epithelium) were the highest expressed pathways. Overall, the IL-1 signaling genes (inflammatory) were relevant in the airway compartment, while pro-Th2 genes including IL-13 and STAT6 were more relevant in fibroblasts, lymphocytes, macrophages and bronchial biopsies. These genes were also associated with asthma in the GWAS catalog. Support Vector Machine showed that DEGs based on macrophages and epithelial cells have the highest and lowest discriminatory accuracy, respectively. Drug (entinostat, BMS-345541) and genetic perturbagens (KLF6, BCL10, INFB1 and BAMBI) negatively connected to disease at multi-tissue level could potentially repurposed for treating asthma. Collectively, our study indicates that the DEGs, perturbagens and disease are connected differentially depending on tissue/cell types. While most of the existing literature describes asthma transcriptome data from individual sample types, the present work demonstrates the utility of multi-tissue transcriptome data. Future studies should focus on collecting transcriptomic data from multiple tissues, age and race groups, genetic background, disease subtypes and on the availability of better-annotated data in the public domain.
Collapse
|
18
|
Alfano DN, Klei LR, Klei HB, Trotta M, Gough PJ, Foley KP, Bertin J, Sumpter TL, Lucas PC, McAllister-Lucas LM. MALT1 Protease Plays a Dual Role in the Allergic Response by Acting in Both Mast Cells and Endothelial Cells. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2020; 204:2337-2348. [PMID: 32213560 DOI: 10.4049/jimmunol.1900281] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/08/2019] [Accepted: 02/21/2020] [Indexed: 01/26/2023]
Abstract
The signaling protein MALT1 plays a key role in promoting NF-κB activation in Ag-stimulated lymphocytes. In this capacity, MALT1 has two functions, acting as a scaffolding protein and as a substrate-specific protease. MALT1 is also required for NF-κB-dependent induction of proinflammatory cytokines after FcεR1 stimulation in mast cells, implicating a role in allergy. Because MALT1 remains understudied in this context, we sought to investigate how MALT1 proteolytic activity contributes to the overall allergic response. We compared bone marrow-derived mast cells from MALT1 knockout (MALT1-/-) and MALT1 protease-deficient (MALTPD/PD) mice to wild-type cells. We found that MALT1-/- and MALT1PD/PD mast cells are equally impaired in cytokine production following FcεRI stimulation, indicating that MALT1 scaffolding activity is insufficient to drive the cytokine response and that MALT1 protease activity is essential. In addition to cytokine production, acute mast cell degranulation is a critical component of allergic response. Intriguingly, whereas degranulation is MALT1-independent, MALT1PD/PD mice are protected from vascular edema induced by either passive cutaneous anaphylaxis or direct challenge with histamine, a major granule component. This suggests a role for MALT1 protease activity in endothelial cells targeted by mast cell-derived vasoactive substances. Indeed, we find that in human endothelial cells, MALT1 protease is activated following histamine treatment and is required for histamine-induced permeability. We thus propose a dual role for MALT1 protease in allergic response, mediating 1) IgE-dependent mast cell cytokine production, and 2) histamine-induced endothelial permeability. This dual role indicates that therapeutic inhibitors of MALT1 protease could work synergistically to control IgE-mediated allergic disease.
Collapse
Affiliation(s)
- Danielle N Alfano
- Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, PA 15224
| | - Linda R Klei
- Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, PA 15224
| | - Hanna B Klei
- Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, PA 15224
| | - Matthew Trotta
- Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, PA 15224
| | - Peter J Gough
- Pattern Recognition Receptor Discovery Performance Unit, GlaxoSmithKline, Collegeville, PA 19406
| | - Kevin P Foley
- Pattern Recognition Receptor Discovery Performance Unit, GlaxoSmithKline, Collegeville, PA 19406
| | - John Bertin
- Pattern Recognition Receptor Discovery Performance Unit, GlaxoSmithKline, Collegeville, PA 19406
| | - Tina L Sumpter
- Department of Dermatology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15224
| | - Peter C Lucas
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15224; and .,Vascular Medicine Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261
| | - Linda M McAllister-Lucas
- Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, PA 15224; .,Vascular Medicine Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261
| |
Collapse
|
19
|
Martin K, Touil R, Cvijetic G, Israel L, Kolb Y, Sarret S, Valeaux S, Degl'Innocenti E, Le Meur T, Caesar N, Bardet M, Beerli C, Zerwes HG, Kovarik J, Beltz K, Schlapbach A, Quancard J, Régnier CH, Bigaud M, Junt T, Wieczorek G, Isnardi I, Littlewood-Evans A, Bornancin F, Calzascia T. Requirement of Mucosa-Associated Lymphoid Tissue Lymphoma Translocation Protein 1 Protease Activity for Fcγ Receptor-Induced Arthritis, but Not Fcγ Receptor-Mediated Platelet Elimination, in Mice. Arthritis Rheumatol 2020; 72:919-930. [PMID: 31943941 DOI: 10.1002/art.41204] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2019] [Accepted: 01/07/2020] [Indexed: 12/11/2022]
Abstract
OBJECTIVE Fcγ receptors (FcγR) play important roles in both protective and pathogenic immune responses. The assembly of the CBM signalosome encompassing caspase recruitment domain-containing protein 9, B cell CLL/lymphoma 10, and mucosa-associated lymphoid tissue lymphoma translocation protein 1 (MALT-1) is required for optimal FcγR-induced canonical NF-κB activation and proinflammatory cytokine release. This study was undertaken to clarify the relevance of MALT-1 protease activity in FcγR-driven events and evaluate the therapeutic potential of selective MALT-1 protease inhibitors in FcγR-mediated diseases. METHODS Using genetic and pharmacologic disruption of MALT-1 scaffolding and enzymatic activity, we assessed the relevance of MALT-1 function in murine and human primary myeloid cells upon stimulation with immune complexes (ICs) and in murine models of autoantibody-driven arthritis and immune thrombocytopenic purpura (ITP). RESULTS MALT-1 protease function is essential for optimal FcγR-induced production of proinflammatory cytokines by various murine and human myeloid cells stimulated with ICs. In contrast, MALT-1 protease inhibition did not affect the Syk-dependent, FcγR-mediated production of reactive oxygen species or leukotriene B4 . Notably, pharmacologic MALT-1 protease inhibition in vivo reduced joint inflammation in the murine K/BxN serum-induced arthritis model (mean area under the curve for paw swelling of 45.42% versus 100% in control mice; P = 0.0007) but did not affect platelet depletion in a passive model of ITP. CONCLUSION Our findings indicate a specific contribution of MALT-1 protease activity to FcγR-mediated events and suggest that MALT-1 protease inhibitors have therapeutic potential in a subset of FcγR-driven inflammatory disorders.
Collapse
Affiliation(s)
- Kea Martin
- Novartis Institutes for BioMedical Research, Basel, Switzerland
| | - Ratiba Touil
- Novartis Institutes for BioMedical Research, Basel, Switzerland
| | | | - Laura Israel
- Novartis Institutes for BioMedical Research, Basel, Switzerland
| | - Yeter Kolb
- Novartis Institutes for BioMedical Research, Basel, Switzerland
| | - Sophie Sarret
- Novartis Institutes for BioMedical Research, Basel, Switzerland
| | | | | | - Thomas Le Meur
- Novartis Institutes for BioMedical Research, Basel, Switzerland
| | - Nadja Caesar
- Novartis Institutes for BioMedical Research, Basel, Switzerland
| | - Maureen Bardet
- Novartis Institutes for BioMedical Research, Basel, Switzerland
| | | | | | - Jiri Kovarik
- Novartis Institutes for BioMedical Research, Basel, Switzerland
| | - Karen Beltz
- Novartis Institutes for BioMedical Research, Basel, Switzerland
| | | | - Jean Quancard
- Novartis Institutes for BioMedical Research, Basel, Switzerland
| | | | - Marc Bigaud
- Novartis Institutes for BioMedical Research, Basel, Switzerland
| | - Tobias Junt
- Novartis Institutes for BioMedical Research, Basel, Switzerland
| | | | | | | | | | | |
Collapse
|
20
|
Phellinus linteus Grown on Germinated Brown Rice Inhibits IgE-Mediated Allergic Activity through the Suppression of Fc εRI-Dependent Signaling Pathway In Vitro and In Vivo. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2019; 2019:1485015. [PMID: 31871471 PMCID: PMC6907041 DOI: 10.1155/2019/1485015] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/05/2019] [Revised: 08/07/2019] [Accepted: 10/17/2019] [Indexed: 12/12/2022]
Abstract
Phellinus linteus (PL) has been used as a traditional herbal medicine owing to its immune regulatory activity. Previous studies reported that PL grown on germinated brown rice (PBR) exerted immunomodulatory, anticancer, and anti-inflammatory activities. However, role of PBR on type I hypersensitive reactions has not been studied yet. We found that PBR contained more polyphenolic compounds than PL extract. Among fractions, PBR butanol fraction (PBR-BuOH) significantly contained the most amounts of total polyphenolic contents compared with all extracts or fractions. In this study, anti-allergic activity of PBR-BuOH was examined using in vitro and in vivo models of immunoglobulin E/antigen- (IgE/Ag-) stimulated allergy. The inhibitory activity of degranulation was higher in PBR-BuOH (IC50 41.31 ± 0.14 μg/mL) than in PL-BuOH (IC50 108.07 ± 8.98 μg/mL). We observed that PBR-BuOH suppressed calcium influx and the level of TNF-α and IL-4 mRNA expression in a dose-dependent manner. The phosphorylation of Fyn, Gab2, PI3K, Syk, and IκB protein is reduced by PBR-BuOH. Oral administration of PBR-BuOH inhibited allergic reactions including the extravasation of Evans blue dye, ear swelling, and infiltration of immune cells in mice with passive cutaneous anaphylaxis (PCA). These findings suggest that PBR-BuOH might be used as a functional food, a health supplement, or a drug for preventing type I hypersensitive allergic disease.
Collapse
|
21
|
Uchida R, Xiang H, Arai H, Kitamura H, Nishida K. L-Type Calcium Channel-Mediated Zinc Wave Is Involved in the Regulation of IL-6 by Stimulating Non-IgE with LPS and IL-33 in Mast Cells and Dendritic Cells. Biol Pharm Bull 2019; 42:87-93. [DOI: 10.1248/bpb.b18-00565] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Affiliation(s)
- Ryota Uchida
- Laboratory of Immune Regulation, Graduate School of Pharmaceutical Sciences, Suzuka University of Medical Science
| | - Huihui Xiang
- Division of Functional Immunology, Institute for Genetic Medicine, Hokkaido University
| | - Hiroya Arai
- Laboratory of Immune Regulation, Faculty of Pharmaceutical Sciences, Suzuka University of Medical Science
| | - Hidemitsu Kitamura
- Division of Functional Immunology, Institute for Genetic Medicine, Hokkaido University
| | - Keigo Nishida
- Laboratory of Immune Regulation, Graduate School of Pharmaceutical Sciences, Suzuka University of Medical Science
- Laboratory of Immune Regulation, Faculty of Pharmaceutical Sciences, Suzuka University of Medical Science
| |
Collapse
|
22
|
Role of Zinc Signaling in the Regulation of Mast Cell-, Basophil-, and T Cell-Mediated Allergic Responses. J Immunol Res 2018; 2018:5749120. [PMID: 30596108 PMCID: PMC6286780 DOI: 10.1155/2018/5749120] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2018] [Accepted: 10/23/2018] [Indexed: 01/26/2023] Open
Abstract
Zinc is essential for maintaining normal structure and physiological function of cells. Its deficiency causes growth retardation, immunodeficiency, and neuronal degeneration. Zinc homeostasis is tightly regulated by zinc transporters and metallothioneins that control zinc concentration and its distribution in individual cells and contributes to zinc signaling. The intracellular zinc signaling regulates immune reactions. Although many molecules involved in these processes have zinc-binding motifs, the molecular mechanisms and the role of zinc in immune responses have not been elucidated. We and others have demonstrated that zinc signaling plays diverse and specific roles in vivo and in vitro in studies using knockout mice lacking zinc transporter function and metallothionein function. In this review, we discuss the impact of zinc signaling focusing particularly on mast cell-, basophil-, and T cell-mediated inflammatory and allergic responses. We also describe zinc signaling dysregulation as a leading health problem in inflammatory disease and allergy.
Collapse
|
23
|
Ruland J, Hartjes L. CARD–BCL-10–MALT1 signalling in protective and pathological immunity. Nat Rev Immunol 2018; 19:118-134. [DOI: 10.1038/s41577-018-0087-2] [Citation(s) in RCA: 90] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
24
|
Juilland M, Thome M. Holding All the CARDs: How MALT1 Controls CARMA/CARD-Dependent Signaling. Front Immunol 2018; 9:1927. [PMID: 30214442 PMCID: PMC6125328 DOI: 10.3389/fimmu.2018.01927] [Citation(s) in RCA: 66] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2018] [Accepted: 08/06/2018] [Indexed: 01/20/2023] Open
Abstract
The scaffold proteins CARMA1-3 (encoded by the genes CARD11, -14 and -10) and CARD9 play major roles in signaling downstream of receptors with immunoreceptor tyrosine activation motifs (ITAMs), G-protein coupled receptors (GPCR) and receptor tyrosine kinases (RTK). These receptors trigger the formation of oligomeric CARMA/CARD-BCL10-MALT1 (CBM) complexes via kinases of the PKC family. The CBM in turn regulates gene expression by the activation of NF-κB and AP-1 transcription factors and controls transcript stability. The paracaspase MALT1 is the only CBM component having an enzymatic (proteolytic) activity and has therefore recently gained attention as a potential drug target. Here we review recent advances in the understanding of the molecular function of the protease MALT1 and summarize how MALT1 scaffold and protease function contribute to the transmission of CBM signals. Finally, we will highlight how dysregulation of MALT1 function can cause pathologies such as immunodeficiency, autoimmunity, psoriasis, and cancer.
Collapse
Affiliation(s)
- Mélanie Juilland
- Department of Biochemistry, University of Lausanne, Epalinges, Switzerland
| | - Margot Thome
- Department of Biochemistry, University of Lausanne, Epalinges, Switzerland
| |
Collapse
|
25
|
Gehring T, Seeholzer T, Krappmann D. BCL10 - Bridging CARDs to Immune Activation. Front Immunol 2018; 9:1539. [PMID: 30022982 PMCID: PMC6039553 DOI: 10.3389/fimmu.2018.01539] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2018] [Accepted: 06/21/2018] [Indexed: 11/25/2022] Open
Abstract
Since the B-cell lymphoma/leukemia 10 (BCL10) protein was first described in 1999, numerous studies have elucidated its key functions in channeling adaptive and innate immune signaling downstream of CARMA/caspase-recruitment domain (CARD) scaffold proteins. While T and B cell antigen receptor (TCR/BCR) signaling induces the recruitment of BCL10 bound to mucosa-associated lymphoid tissue (MALT)1 to the lymphocyte-specific CARMA1/CARD11–BCL10–MALT1 (CBM-1) signalosome, alternative CBM complexes utilize different CARMA/CARD scaffolds in distinct innate or inflammatory pathways. BCL10 constitutes the smallest subunit in all CBM signalosomes, containing a 233 amino acid coding for N-terminal CARD as well as a C-terminal Ser/Thr-rich region. BCL10 forms filaments, thereby aggregating into higher-order clusters that mediate and amplify stimulation-induced signals, ultimately leading to MALT1 protease activation and canonical NF-κB and JNK signaling. BCL10 additionally undergoes extensive post-translational regulation involving phosphorylation, ubiquitination, MALT1-catalyzed cleavage, and degradation. Through these feedback and feed-forward events, BCL10 integrates positive and negative regulatory processes that govern the function as well as the dynamic assembly, disassembly, and destruction of CBM complexes. Thus, BCL10 is a critical regulator for activation as well as termination of immune cell signaling, revealing that its role extends far beyond that of a mere linking factor in CBM complexes.
Collapse
Affiliation(s)
- Torben Gehring
- Research Unit Cellular Signal Integration, Institute of Molecular Toxicology and Pharmacology, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany
| | - Thomas Seeholzer
- Research Unit Cellular Signal Integration, Institute of Molecular Toxicology and Pharmacology, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany
| | - Daniel Krappmann
- Research Unit Cellular Signal Integration, Institute of Molecular Toxicology and Pharmacology, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany
| |
Collapse
|
26
|
Castillo-Arellano JI, Guzmán-Gutiérrez SL, Ibarra-Sánchez A, Hernández-Ortega S, Nieto-Camacho A, Medina-Campos ON, Pedraza-Chaverri J, Reyes-Chilpa R, González-Espinosa C. Jacareubin inhibits FcεRI-induced extracellular calcium entry and production of reactive oxygen species required for anaphylactic degranulation of mast cells. Biochem Pharmacol 2018; 154:344-356. [PMID: 29802828 DOI: 10.1016/j.bcp.2018.05.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2018] [Accepted: 05/03/2018] [Indexed: 12/27/2022]
Abstract
Mast cells (MCs) are important effectors in allergic reactions since they produce a number of pre-formed and de novo synthesized pro-inflammatory compounds in response to the high affinity IgE receptor (FcεRI) crosslinking. IgE/Antigen-dependent degranulation and cytokine synthesis in MCs have been recognized as relevant pharmacological targets for the control of deleterious inflammatory reactions. Despite the relevance of allergic diseases worldwide, efficient pharmacological control of mast cell degranulation has been elusive. In this work, the xanthone jacareubin was isolated from the heartwood of the tropical tree Callophyllum brasilense, and its tridimensional structure was determined for the first time by X-ray diffraction. Also, its effects on the main activation parameters of bone marrow-derived mast cells (BMMCs) were evaluated. Jacareubin inhibited IgE/Ag-induced degranulation in a dose-response manner with an IC50 = 46 nM. It also blocked extracellular calcium influx triggered by IgE/Ag complexes and by the SERCA ATPase inhibitor thapsigargin (Thap). Inhibition of calcium entry correlated with a blockage on the reactive oxygen species (ROS) accumulation. Antioxidant capacity of jacareubin was higher than the showed by α-tocopherol and caffeic acid, but similar to trolox. Jacareubin shown inhibitory actions on xanthine oxidase, but not on NADPH oxidase (NOX) activities. In vivo, jacareubin inhibited passive anaphylactic reactions and TPA-induced edema in mice. Our data demonstrate that jacareubin is a potent natural compound able to inhibit anaphylactic degranualtion in mast cells by blunting FcεRI-induced calcium flux needed for secretion of granule content, and suggest that xanthones could be efficient anti-oxidant, antiallergic, and antiinflammatory molecules.
Collapse
Affiliation(s)
- J I Castillo-Arellano
- Departamento de Farmacobiología, Centro de Investigación y de Estudios Avanzados del IPN, Mexico; Instituto de Química, Universidad Nacional Autónoma de México, Mexico
| | - S L Guzmán-Gutiérrez
- Departamento de Inmunología, Catedrática CONACyT-Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mexico
| | - A Ibarra-Sánchez
- Departamento de Farmacobiología, Centro de Investigación y de Estudios Avanzados del IPN, Mexico
| | | | - A Nieto-Camacho
- Instituto de Química, Universidad Nacional Autónoma de México, Mexico
| | - O N Medina-Campos
- Departamento de Bioquímica, Facultad de Química, Universidad Nacional Autónoma de México, Mexico
| | - J Pedraza-Chaverri
- Departamento de Bioquímica, Facultad de Química, Universidad Nacional Autónoma de México, Mexico
| | - R Reyes-Chilpa
- Instituto de Química, Universidad Nacional Autónoma de México, Mexico.
| | - C González-Espinosa
- Departamento de Farmacobiología, Centro de Investigación y de Estudios Avanzados del IPN, Mexico.
| |
Collapse
|
27
|
Park YH, Kim DK, Kim HW, Kim HS, Lee D, Lee MB, Min KY, Koo J, Kim SJ, Kang C, Kim YM, Kim HS, Choi WS. Repositioning of anti-cancer drug candidate, AZD7762, to an anti-allergic drug suppressing IgE-mediated mast cells and allergic responses via the inhibition of Lyn and Fyn. Biochem Pharmacol 2018; 154:270-277. [PMID: 29777684 DOI: 10.1016/j.bcp.2018.05.012] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2018] [Accepted: 05/15/2018] [Indexed: 01/26/2023]
Abstract
Mast cells are critical effector cells in IgE-mediated allergic responses. The aim of this study was to investigate the anti-allergic effects of 3-[(aminocarbonyl)amino]-5-(3-fluorophenyl)-N-(3S)-3-piperidinyl-2-thiophenecarboxamide (AZD7762) in vitro and in vivo. AZD7762 inhibited the antigen-stimulated degranulation from RBL-2H3 (IC50, ∼27.9 nM) and BMMCs (IC50, ∼99.3 nM) in a dose-dependent manner. AZD7762 also inhibited the production of TNF-α and IL-4. As the mechanism of its action, AZD7762 inhibited the activation of Syk and its downstream signaling proteins, such as Linker of activated T cells (LAT), phospholipase (PL) Cγ1, Akt, and mitogen-activated protein (MAP) kinases (Erk1/2, p38, and JNK) in mast cells. In in vitro protein kinase assay, AZD7762 inhibited the activity of Lyn and Fyn kinases, which are important for the activation of Syk in mast cells. Furthermore, AZD7762 also suppressed the degranulation of LAD2 human mast cells (IC50, ∼49.9 nM) and activation of Syk in a dose-dependent manner. As observed in experiments with mast cells in vitro, AZD7762 inhibited antigen-mediated passive cutaneous anaphylaxis in mice (ED50, ∼35.8 mg/kg). Altogether, these results suggest that AZD7762 could be used as a new therapeutic agent for mast cell-mediated allergic diseases.
Collapse
Affiliation(s)
- Young Hwan Park
- Department of Immunology, College of Medicine, Konkuk University, Chungju 27478, Republic of Korea
| | - Do-Kyun Kim
- Mast Cell Biology Section, Laboratory of Allergic Diseases, National Institute of Allergy and Infectious Diseases, Bethesda, MD 20892, USA
| | - Hyun Woo Kim
- Department of Immunology, College of Medicine, Konkuk University, Chungju 27478, Republic of Korea
| | - Hyuk Soon Kim
- Department of Immunology, College of Medicine, Konkuk University, Chungju 27478, Republic of Korea
| | - Dajeong Lee
- Department of Immunology, College of Medicine, Konkuk University, Chungju 27478, Republic of Korea
| | - Min Bum Lee
- Department of Immunology, College of Medicine, Konkuk University, Chungju 27478, Republic of Korea
| | - Keun Young Min
- Department of Immunology, College of Medicine, Konkuk University, Chungju 27478, Republic of Korea
| | - Jimo Koo
- Department of Immunology, College of Medicine, Konkuk University, Chungju 27478, Republic of Korea
| | - Su Jeong Kim
- Department of Immunology, College of Medicine, Konkuk University, Chungju 27478, Republic of Korea
| | - Changhee Kang
- Department of Stem Cell Biology, School of Medicine, Konkuk University, Seoul 05029, Republic of Korea
| | - Young Mi Kim
- College of Pharmacy, Duksung Women's University, Seoul 01369, Republic of Korea
| | - Hyung Sik Kim
- Division of Toxicology, College of Pharmacy, Sungkyunkwan University, Suwon, Gyeonggi-do 16419, Republic of Korea
| | - Wahn Soo Choi
- Department of Immunology, College of Medicine, Konkuk University, Chungju 27478, Republic of Korea.
| |
Collapse
|
28
|
Poplutz M, Levikova M, Lüscher-Firzlaff J, Lesina M, Algül H, Lüscher B, Huber M. Endotoxin tolerance in mast cells, its consequences for IgE-mediated signalling, and the effects of BCL3 deficiency. Sci Rep 2017; 7:4534. [PMID: 28674400 PMCID: PMC5495797 DOI: 10.1038/s41598-017-04890-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2017] [Accepted: 05/30/2017] [Indexed: 11/30/2022] Open
Abstract
Stimulation with lipopolysaccharide (LPS; endotoxin) not only causes rapid production of proinflammatory cytokines, but also induces a state of LPS hypo-responsiveness to a second LPS stimulation (endotoxin tolerance (ET)). Murine bone marrow-derived MCs (BMMCs) and peritoneal MCs (PMCs) developed ET as shown by an abrogated production of Il6/Tnf RNAs and IL-6/TNF-α proteins. In naive BMMCs, LPS stimulation induced a transient decline in the trimethylation of lysine 9 of the core histone H3 (H3K9me3), a suppressive chromatin mark, at the Il6/Tnf promoters, which correlated with p50(NFκB) and p65(NFκB) binding. Both demethylation and NFκB binding were abrogated in tolerant cells. In addition, cytosolic NFκB activation was suppressed in tolerant BMMCs. Intriguingly, antigen stimulation of naive and tolerant MCs induced comparable production of Il6/Tnf and IL-6/TNF-α, although ET also affected antigen-triggered activation of NFκB; pharmacological analysis indicated the importance of Ca2+-dependent transcription in this respect. In macrophages, the IκB member BCL3 is induced by LPS and known to be involved in ET, which was not corroborated comparing wild-type and Bcl3-deficient BMMCs. Interestingly, Bcl3-deficient PMCs produce markedly increased amounts of IL-6/TNF-α after LPS stimulation. Collectively, ET in MCs is BCL3-independent, however, in PMCs, BCL3 negatively regulates immediate LPS-induced cytokine production and quantitatively affects ET.
Collapse
Affiliation(s)
- Magdalena Poplutz
- Institute of Biochemistry and Molecular Immunology, Medical School, RWTH Aachen University, Aachen, Germany
| | - Maryna Levikova
- Institute of Biochemistry and Molecular Biology, Medical School, RWTH Aachen University, Aachen, Germany
- Institute of Molecular Cancer Research, University of Zürich, Zürich, Switzerland
| | - Juliane Lüscher-Firzlaff
- Institute of Biochemistry and Molecular Biology, Medical School, RWTH Aachen University, Aachen, Germany
| | - Marina Lesina
- Molecular Gastroenterology, Medical Clinic II, University Hospital Klinikum Rechts der Isar, TU Munich, Munich, Germany
| | - Hana Algül
- Molecular Gastroenterology, Medical Clinic II, University Hospital Klinikum Rechts der Isar, TU Munich, Munich, Germany
| | - Bernhard Lüscher
- Institute of Biochemistry and Molecular Biology, Medical School, RWTH Aachen University, Aachen, Germany
| | - Michael Huber
- Institute of Biochemistry and Molecular Immunology, Medical School, RWTH Aachen University, Aachen, Germany.
| |
Collapse
|
29
|
Nunes de Miranda SM, Wilhelm T, Huber M, Zorn CN. Differential Lyn-dependence of the SHIP1-deficient mast cell phenotype. Cell Commun Signal 2016; 14:12. [PMID: 27206658 PMCID: PMC4874025 DOI: 10.1186/s12964-016-0135-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2015] [Accepted: 05/13/2016] [Indexed: 01/02/2023] Open
Abstract
Background Antigen (Ag)/IgE-mediated mast cell (MC) responses play detrimental roles in allergic diseases. MC activation via the high-affinity receptor for IgE (FcεRI) is controlled by the Src family kinase Lyn. Lyn-deficient (-/-) bone marrow-derived MCs (BMMCs) have been shown by various laboratories to exert stronger activation of the PI3K pathway, degranulation, and production of pro-inflammatory cytokines compared to wild-type (wt) cells. This mimics the phenotype of BMMCs deficient for the SH2-containing inositol-5’-phosphatase 1 (SHIP1). In this line, Lyn has been demonstrated to tyrosine-phosphorylate and activate SHIP1, thereby constituting a negative feedback control of PI3K-mediated signals. However, several groups have also reported on Lyn-/- BMMCs degranulating weaker than wt BMMCs. Results Lyn-/- BMMCs, which show a suppressed degranulation response, were found to exhibit abrogated tyrosine phosphorylation of SHIP1 as well. This indicated that even in the presence of reduced SHIP1 function MC degranulation is dependent on Lyn function. In contrast to the reduced immediate secretory response, pro-inflammatory cytokine production was augmented in Lyn-/- BMMCs. For closer analysis, Lyn/SHIP1-double-deficient (dko) BMMCs were generated. In support of the dominance of Lyn deficiency, dko BMMCs degranulated significantly weaker than SHIP1-/- BMMCs. This coincided with reduced LAT1 and PLC-γ1 phosphorylation as well as Ca2+ mobilization in those cells. Interestingly, activation of the NFκB pathway followed the same pattern as measured by IκBα phosphorylation/degradation as well as induction of NFκB target genes. This suggested that Ag-triggered NFκB activation involves a Ca2+-dependent step. Indeed, IκBα phosphorylation/degradation and NFκB target gene induction were controlled by the Ca2+-dependent phosphatase calcineurin. Conclusions Lyn deficiency is dominant over SHIP1 deficiency in MCs with respect to Ag-triggered degranulation and preceding signaling events. Moreover, the NFκB pathway and respective targets are activated in a Lyn- and Ca2+-dependent manner, reinforcing the importance of Lyn for MC activation. Electronic supplementary material The online version of this article (doi:10.1186/s12964-016-0135-0) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Susana M Nunes de Miranda
- Institute of Biochemistry and Molecular Immunology, University Clinic, RWTH Aachen University, Pauwelsstraße 30, Aachen, 52074, Germany
| | - Thomas Wilhelm
- Institute of Biochemistry and Molecular Immunology, University Clinic, RWTH Aachen University, Pauwelsstraße 30, Aachen, 52074, Germany
| | - Michael Huber
- Institute of Biochemistry and Molecular Immunology, University Clinic, RWTH Aachen University, Pauwelsstraße 30, Aachen, 52074, Germany.
| | - Carolin N Zorn
- Institute of Biochemistry and Molecular Immunology, University Clinic, RWTH Aachen University, Pauwelsstraße 30, Aachen, 52074, Germany
| |
Collapse
|
30
|
Martín-Ávila A, Medina-Tamayo J, Ibarra-Sánchez A, Vázquez-Victorio G, Castillo-Arellano JI, Hernández-Mondragón AC, Rivera J, Madera-Salcedo IK, Blank U, Macías-Silva M, González-Espinosa C. Protein Tyrosine Kinase Fyn Regulates TLR4-Elicited Responses on Mast Cells Controlling the Function of a PP2A-PKCα/β Signaling Node Leading to TNF Secretion. THE JOURNAL OF IMMUNOLOGY 2016; 196:5075-88. [PMID: 27183589 DOI: 10.4049/jimmunol.1501823] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/13/2015] [Accepted: 04/16/2016] [Indexed: 12/21/2022]
Abstract
Mast cells produce proinflammatory cytokines in response to TLR4 ligands, but the signaling pathways involved are not fully described. In this study, the participation of the Src family kinase Fyn in the production of TNF after stimulation with LPS was evaluated using bone marrow-derived mast cells from wild-type and Fyn-deficient mice. Fyn(-/-) cells showed higher LPS-induced secretion of preformed and de novo-synthesized TNF. In both cell types, TNF colocalized with vesicle-associated membrane protein (VAMP)3-positive compartments. Addition of LPS provoked coalescence of VAMP3 and its interaction with synaptosomal-associated protein 23; those events were increased in the absence of Fyn. Higher TNF mRNA levels were also observed in Fyn-deficient cells as a result of increased transcription and greater mRNA stability after LPS treatment. Fyn(-/-) cells also showed higher LPS-induced activation of TAK-1 and ERK1/2, whereas IκB kinase and IκB were phosphorylated, even in basal conditions. Increased responsiveness in Fyn(-/-) cells was associated with a lower activity of protein phosphatase 2A (PP2A) and augmented activity of protein kinase C (PKC)α/β, which was dissociated from PP2A and increased its association with the adapter protein neuroblast differentiation-associated protein (AHNAK, desmoyokin). LPS-induced PKCα/β activity was associated with VAMP3 coalescence in WT and Fyn-deficient cells. Reconstitution of MC-deficient Wsh mice with Fyn(-/-) MCs produced greater LPS-dependent production of TNF in the peritoneal cavity. Our data show that Fyn kinase is activated after TLR4 triggering and exerts an important negative control on LPS-dependent TNF production in MCs controlling the inactivation of PP2Ac and activation of PKCα/β necessary for the secretion of TNF by VAMP3(+) carriers.
Collapse
Affiliation(s)
- Alejandro Martín-Ávila
- Departamento de Farmacobiología, Centro de Investigación y Estudios Avanzados del Instituto Politécnico Nacional, Sede Sur, Tlalpan, CP 14330 Mexico City, Mexico
| | - Jaciel Medina-Tamayo
- Departamento de Farmacobiología, Centro de Investigación y Estudios Avanzados del Instituto Politécnico Nacional, Sede Sur, Tlalpan, CP 14330 Mexico City, Mexico
| | - Alfredo Ibarra-Sánchez
- Departamento de Farmacobiología, Centro de Investigación y Estudios Avanzados del Instituto Politécnico Nacional, Sede Sur, Tlalpan, CP 14330 Mexico City, Mexico
| | - Genaro Vázquez-Victorio
- Departamento de Biología Celular y del Desarrollo, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Ciudad Universitaria, CP 04510 Mexico City, Mexico
| | - Jorge Iván Castillo-Arellano
- Departamento de Farmacobiología, Centro de Investigación y Estudios Avanzados del Instituto Politécnico Nacional, Sede Sur, Tlalpan, CP 14330 Mexico City, Mexico
| | - Alma Cristal Hernández-Mondragón
- Departamento de Farmacobiología, Centro de Investigación y Estudios Avanzados del Instituto Politécnico Nacional, Sede Sur, Tlalpan, CP 14330 Mexico City, Mexico
| | - Juan Rivera
- National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD 20892-1820; and
| | - Iris K Madera-Salcedo
- INSERM UMRS1149, Faculté de Médecine, Université Paris-Diderot, Site X, Bichat, Paris 75018, France
| | - Ulrich Blank
- INSERM UMRS1149, Faculté de Médecine, Université Paris-Diderot, Site X, Bichat, Paris 75018, France
| | - Marina Macías-Silva
- Departamento de Biología Celular y del Desarrollo, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Ciudad Universitaria, CP 04510 Mexico City, Mexico
| | - Claudia González-Espinosa
- Departamento de Farmacobiología, Centro de Investigación y Estudios Avanzados del Instituto Politécnico Nacional, Sede Sur, Tlalpan, CP 14330 Mexico City, Mexico;
| |
Collapse
|
31
|
Bugajev V, Halova I, Draberova L, Bambouskova M, Potuckova L, Draberova H, Paulenda T, Junyent S, Draber P. Negative regulatory roles of ORMDL3 in the FcεRI-triggered expression of proinflammatory mediators and chemotactic response in murine mast cells. Cell Mol Life Sci 2016; 73:1265-85. [PMID: 26407610 PMCID: PMC11108389 DOI: 10.1007/s00018-015-2047-3] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2015] [Revised: 08/19/2015] [Accepted: 09/17/2015] [Indexed: 10/23/2022]
Abstract
Single-nucleotide polymorphism studies have linked the chromosome 17q12-q21 region, where the human orosomucoid-like (ORMDL)3 gene is localized, to the risk of asthma and several other inflammatory diseases. Although mast cells are involved in the development of these diseases, the contribution of ORMDL3 to the mast cell physiology is unknown. In this study, we examined the role of ORMDL3 in antigen-induced activation of murine mast cells with reduced or enhanced ORMDL3 expression. Our data show that in antigen-activated mast cells, reduced expression of the ORMDL3 protein had no effect on degranulation and calcium response, but significantly enhanced phosphorylation of AKT kinase at Ser 473 followed by enhanced phosphorylation and degradation of IκBα and translocation of the NF-κB p65 subunit into the nucleus. These events were associated with an increased expression of proinflammatory cytokines (TNF-α, IL-6, and IL-13), chemokines (CCL3 and CCL4), and cyclooxygenase-2 dependent synthesis of prostaglandin D2. Antigen-mediated chemotaxis was also enhanced in ORMDL3-deficient cells, whereas spreading on fibronectin was decreased. On the other hand, increased expression of ORMDL3 had no significant effect on the studied signaling events, except for reduced antigen-mediated chemotaxis. These data were corroborated by increased IgE-antigen-dependent passive cutaneous anaphylaxis in mice with locally silenced ORMDL3 using short interfering RNAs. Our data also show that antigen triggers suppression of ORMDL3 expression in the mast cells. In summary, we provide evidence that downregulation of ORMDL3 expression in mast cells enhances AKT and NF-κB-directed signaling pathways and chemotaxis and contributes to the development of mast cell-mediated local inflammation in vivo.
Collapse
Affiliation(s)
- Viktor Bugajev
- Department of Signal Transduction, Institute of Molecular Genetics, Academy of Sciences of the Czech Republic, v.v.i., Videnska 1083, 142 20, Prague 4, Czech Republic
| | - Ivana Halova
- Department of Signal Transduction, Institute of Molecular Genetics, Academy of Sciences of the Czech Republic, v.v.i., Videnska 1083, 142 20, Prague 4, Czech Republic
| | - Lubica Draberova
- Department of Signal Transduction, Institute of Molecular Genetics, Academy of Sciences of the Czech Republic, v.v.i., Videnska 1083, 142 20, Prague 4, Czech Republic
| | - Monika Bambouskova
- Department of Signal Transduction, Institute of Molecular Genetics, Academy of Sciences of the Czech Republic, v.v.i., Videnska 1083, 142 20, Prague 4, Czech Republic
| | - Lucie Potuckova
- Department of Signal Transduction, Institute of Molecular Genetics, Academy of Sciences of the Czech Republic, v.v.i., Videnska 1083, 142 20, Prague 4, Czech Republic
| | - Helena Draberova
- Department of Signal Transduction, Institute of Molecular Genetics, Academy of Sciences of the Czech Republic, v.v.i., Videnska 1083, 142 20, Prague 4, Czech Republic
| | - Tomas Paulenda
- Department of Signal Transduction, Institute of Molecular Genetics, Academy of Sciences of the Czech Republic, v.v.i., Videnska 1083, 142 20, Prague 4, Czech Republic
| | - Sergi Junyent
- Department of Signal Transduction, Institute of Molecular Genetics, Academy of Sciences of the Czech Republic, v.v.i., Videnska 1083, 142 20, Prague 4, Czech Republic
| | - Petr Draber
- Department of Signal Transduction, Institute of Molecular Genetics, Academy of Sciences of the Czech Republic, v.v.i., Videnska 1083, 142 20, Prague 4, Czech Republic.
| |
Collapse
|
32
|
Abstract
Polymorphisms in the T cell (or transmembrane) immunoglobulin and mucin
domain 1 ( TIM-1) gene, particularly in the mucin
domain, have been associated with atopy and allergic diseases in mice and human.
Genetic- and antibody-mediated studies revealed that Tim-1 functions as a
positive regulator of Th2 responses, while certain antibodies to Tim-1 can
exacerbate or reduce allergic lung inflammation. Tim-1 can also positively
regulate the function of B cells, NKT cells, dendritic cells and mast cells.
However, the precise molecular mechanisms by which Tim-1 modulates immune cell
function are currently unknown. In this study, we have focused on defining
Tim-1-mediated signaling pathways that enhance mast cell activation through the
high affinity IgE receptor (FceRI). Using a Tim-1 mouse model lacking the mucin
domain (Tim-1 Dmucin), we show for the first time that the
polymorphic Tim-1 mucin region is dispensable for normal mast cell activation.
We further show that Tim-4 cross-linking of Tim-1 enhances select signaling
pathways downstream of FceRI in mast cells, including mTOR-dependent signaling,
leading to increased cytokine production but without affecting
degranulation.
Collapse
Affiliation(s)
- Binh Phong
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, USA.,Immunology Graduate Program, University of Pittsburgh School of Medicine, Pittsburgh, USA
| | - Lawrence P Kane
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, USA
| |
Collapse
|
33
|
Jaworski M, Thome M. The paracaspase MALT1: biological function and potential for therapeutic inhibition. Cell Mol Life Sci 2016; 73:459-73. [PMID: 26507244 PMCID: PMC4713714 DOI: 10.1007/s00018-015-2059-z] [Citation(s) in RCA: 84] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2015] [Revised: 09/15/2015] [Accepted: 09/29/2015] [Indexed: 02/07/2023]
Abstract
The paracaspase MALT1 has a central role in the activation of lymphocytes and other immune cells including myeloid cells, mast cells and NK cells. MALT1 activity is required not only for the immune response, but also for the development of natural Treg cells that keep the immune response in check. Exaggerated MALT1 activity has been associated with the development of lymphoid malignancies, and recently developed MALT1 inhibitors show promising anti-tumor effects in xenograft models of diffuse large B cell lymphoma. In this review, we provide an overview of the present understanding of MALT1's function, and discuss possibilities for its therapeutic targeting based on recently developed inhibitors and animal models.
Collapse
Affiliation(s)
- Maike Jaworski
- Department of Biochemistry, University of Lausanne, 1066, Epalinges, Switzerland
| | - Margot Thome
- Department of Biochemistry, University of Lausanne, 1066, Epalinges, Switzerland.
| |
Collapse
|
34
|
Drube S, Weber F, Loschinski R, Beyer M, Rothe M, Rabenhorst A, Göpfert C, Meininger I, Diamanti MA, Stegner D, Häfner N, Böttcher M, Reinecke K, Herdegen T, Greten FR, Nieswandt B, Hartmann K, Krämer OH, Kamradt T. Subthreshold IKK activation modulates the effector functions of primary mast cells and allows specific targeting of transformed mast cells. Oncotarget 2016; 6:5354-68. [PMID: 25749030 PMCID: PMC4467154 DOI: 10.18632/oncotarget.3022] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2014] [Accepted: 12/31/2014] [Indexed: 01/16/2023] Open
Abstract
Mast cell differentiation and proliferation depends on IL-3. IL-3 induces the activation of MAP-kinases and STATs and consequently induces proliferation and survival. Dysregulation of IL-3 signaling pathways also contribute to inflammation and tumorigenesis. We show here that IL-3 induces a SFK- and Ca²⁺-dependent activation of the inhibitor of κB kinases 2 (IKK2) which results in mast cell proliferation and survival but does not induce IκBα-degradation and NFκB activation. Therefore we propose the term "subthreshold IKK activation".This subthreshold IKK activation also primes mast cells for enhanced responsiveness to IL-33R signaling. Consequently, co-stimulation with IL-3 and IL-33 increases IKK activation and massively enhances cytokine production induced by IL-33.We further reveal that in neoplastic mast cells expressing constitutively active Ras, subthreshold IKK activation is associated with uncontrolled proliferation. Consequently, pharmacological IKK inhibition reduces tumor growth selectively by inducing apoptosis in vivo.Together, subthreshold IKK activation is crucial to mediate the full IL-33-induced effector functions in primary mast cells and to mediate uncontrolled proliferation of neoplastic mast cells. Thus, IKK2 is a new molecularly defined target structure.
Collapse
Affiliation(s)
- Sebastian Drube
- Institut für Immunologie, Universitätsklinikum Jena, 07743 Jena, Germany
| | - Franziska Weber
- Institut für Immunologie, Universitätsklinikum Jena, 07743 Jena, Germany
| | - Romy Loschinski
- Institut für Immunologie, Universitätsklinikum Jena, 07743 Jena, Germany
| | - Mandy Beyer
- Institut für Immunologie, Universitätsklinikum Jena, 07743 Jena, Germany
| | - Mandy Rothe
- Institut für Immunologie, Universitätsklinikum Jena, 07743 Jena, Germany
| | - Anja Rabenhorst
- Klinik und Poliklinik für Dermatologie und Venerologie, Universität zu Köln, 50937 Köln, Germany
| | - Christiane Göpfert
- Institut für Immunologie, Universitätsklinikum Jena, 07743 Jena, Germany
| | - Isabel Meininger
- Institut für Immunologie, Universitätsklinikum Jena, 07743 Jena, Germany
| | - Michaela A Diamanti
- Georg-Speyer-Haus, Institute for Tumorbiology and Experimental Therapy, 60596 Frankfurt, Germany
| | - David Stegner
- Rudolf Virchow Centrum für experimentelle Biomedizin, Universität Würzburg, 97080 Würzburg, Germany
| | - Norman Häfner
- Gynäkologische Molekularbiologie, Klinik für Frauenheilkunde und Geburtshilfe, 07743 Jena, Germany
| | - Martin Böttcher
- Institut für Immunologie, Universitätsklinikum Jena, 07743 Jena, Germany
| | - Kirstin Reinecke
- Institut für Experimentelle und Klinische Pharmakologie, Universität Schleswig-Holstein, 24105 Kiel, Germany
| | - Thomas Herdegen
- Institut für Experimentelle und Klinische Pharmakologie, Universität Schleswig-Holstein, 24105 Kiel, Germany
| | - Florian R Greten
- Georg-Speyer-Haus, Institute for Tumorbiology and Experimental Therapy, 60596 Frankfurt, Germany
| | - Bernhard Nieswandt
- Rudolf Virchow Centrum für experimentelle Biomedizin, Universität Würzburg, 97080 Würzburg, Germany
| | - Karin Hartmann
- Klinik und Poliklinik für Dermatologie und Venerologie, Universität zu Köln, 50937 Köln, Germany
| | - Oliver H Krämer
- Institut für Toxikologie, Universitätsmedizin Mainz, 55131 Mainz, Germany
| | - Thomas Kamradt
- Institut für Immunologie, Universitätsklinikum Jena, 07743 Jena, Germany
| |
Collapse
|
35
|
Demeyer A, Staal J, Beyaert R. Targeting MALT1 Proteolytic Activity in Immunity, Inflammation and Disease: Good or Bad? Trends Mol Med 2016; 22:135-150. [PMID: 26787500 DOI: 10.1016/j.molmed.2015.12.004] [Citation(s) in RCA: 57] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2015] [Revised: 12/15/2015] [Accepted: 12/16/2015] [Indexed: 01/05/2023]
Abstract
MALT1 is a signaling protein that plays a key role in immunity, inflammation, and lymphoid malignancies. For a long time MALT1 was believed to function as a scaffold protein, providing an assembly platform for other signaling proteins. This view changed dramatically when MALT1 was also found to have proteolytic activity and a capacity to fine-tune immune responses. Preclinical studies have fostered the belief that MALT1 is a promising therapeutic target in autoimmunity and B cell lymphomas. However, recent studies have shown that mice expressing catalytically-inactive MALT1 develop multi-organ inflammation and autoimmunity, and thus have tempered this initial enthusiasm. We discuss recent findings, highlighting the urgent need for a better mechanistic and functional understanding of MALT1 in host defense and disease.
Collapse
Affiliation(s)
- Annelies Demeyer
- Inflammation Research Center, Unit of Molecular Signal Transduction in Inflammation, Vlaams Instituut voor Biotechnologie (VIB), 9052 Ghent, Belgium; Department of Biomedical Molecular Biology, Ghent University, 9052 Ghent, Belgium
| | - Jens Staal
- Inflammation Research Center, Unit of Molecular Signal Transduction in Inflammation, Vlaams Instituut voor Biotechnologie (VIB), 9052 Ghent, Belgium; Department of Biomedical Molecular Biology, Ghent University, 9052 Ghent, Belgium
| | - Rudi Beyaert
- Inflammation Research Center, Unit of Molecular Signal Transduction in Inflammation, Vlaams Instituut voor Biotechnologie (VIB), 9052 Ghent, Belgium; Department of Biomedical Molecular Biology, Ghent University, 9052 Ghent, Belgium.
| |
Collapse
|
36
|
Abstract
The human paracaspase MALT1 is a caspase homolog that plays a central role in NF-κB signaling. Over the past few years it has become clear that this is due to a combination of its scaffolding and proteolytic function. Knockout mice and mice expressing a catalytically dead variant of the protease have provided valuable information. This review aims to provide an overview of recent developments regarding the enzymatic mechanism and specificity of MALT1, its substrates discovered to date, different mouse models, as well as the role of MALT1 in NF-κB signaling downstream of a variety of different receptors.
Collapse
|
37
|
Yu JW, Hoffman S, Beal AM, Dykon A, Ringenberg MA, Hughes AC, Dare L, Anderson AD, Finger J, Kasparcova V, Rickard D, Berger SB, Ramanjulu J, Emery JG, Gough PJ, Bertin J, Foley KP. MALT1 Protease Activity Is Required for Innate and Adaptive Immune Responses. PLoS One 2015; 10:e0127083. [PMID: 25965667 PMCID: PMC4428694 DOI: 10.1371/journal.pone.0127083] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2014] [Accepted: 04/11/2015] [Indexed: 11/26/2022] Open
Abstract
CARMA-BCL10-MALT1 signalosomes play important roles in antigen receptor signaling and other pathways. Previous studies have suggested that as part of this complex, MALT1 functions as both a scaffolding protein to activate NF-κB through recruitment of ubiquitin ligases, and as a protease to cleave and inactivate downstream inhibitory signaling proteins. However, our understanding of the relative importance of these two distinct MALT1 activities has been hampered by a lack of selective MALT1 protease inhibitors with suitable pharmacologic properties. To fully investigate the role of MALT1 protease activity, we generated mice homozygous for a protease-dead mutation in MALT1. We found that some, but not all, MALT1 functions in immune cells were dependent upon its protease activity. Protease-dead mice had defects in the generation of splenic marginal zone and peritoneal B1 B cells. CD4+ and CD8+ T cells displayed decreased T cell receptor-stimulated proliferation and IL-2 production while B cell receptor-stimulated proliferation was partially dependent on protease activity. In dendritic cells, stimulation of cytokine production through the Dectin-1, Dectin-2, and Mincle C-type lectin receptors was also found to be partially dependent upon protease activity. In vivo, protease-dead mice had reduced basal immunoglobulin levels, and showed defective responses to immunization with T-dependent and T-independent antigens. Surprisingly, despite these decreased responses, MALT1 protease-dead mice, but not MALT1 null mice, developed mixed inflammatory cell infiltrates in multiple organs, suggesting MALT1 protease activity plays a role in immune homeostasis. These findings highlight the importance of MALT1 protease activity in multiple immune cell types, and in integrating immune responses in vivo.
Collapse
Affiliation(s)
- Jong W. Yu
- Pattern Recognition Receptor Discovery Performance Unit, Immuno-Inflammation Therapeutic Area, GlaxoSmithKline, Collegeville, United States of America
| | - Sandy Hoffman
- Pattern Recognition Receptor Discovery Performance Unit, Immuno-Inflammation Therapeutic Area, GlaxoSmithKline, Collegeville, United States of America
| | - Allison M. Beal
- Pattern Recognition Receptor Discovery Performance Unit, Immuno-Inflammation Therapeutic Area, GlaxoSmithKline, Collegeville, United States of America
| | - Angela Dykon
- Pattern Recognition Receptor Discovery Performance Unit, Immuno-Inflammation Therapeutic Area, GlaxoSmithKline, Collegeville, United States of America
| | - Michael A. Ringenberg
- Department of Safety Assessment, GlaxoSmithKline, King of Prussia, United States of America
| | - Anna C. Hughes
- Department of Safety Assessment, GlaxoSmithKline, King of Prussia, United States of America
| | - Lauren Dare
- Pattern Recognition Receptor Discovery Performance Unit, Immuno-Inflammation Therapeutic Area, GlaxoSmithKline, Collegeville, United States of America
| | - Amber D. Anderson
- Quantitative Sciences, GlaxoSmithKline, Collegeville, United States of America
| | - Joshua Finger
- Pattern Recognition Receptor Discovery Performance Unit, Immuno-Inflammation Therapeutic Area, GlaxoSmithKline, Collegeville, United States of America
| | - Viera Kasparcova
- Pattern Recognition Receptor Discovery Performance Unit, Immuno-Inflammation Therapeutic Area, GlaxoSmithKline, Collegeville, United States of America
| | - David Rickard
- Pattern Recognition Receptor Discovery Performance Unit, Immuno-Inflammation Therapeutic Area, GlaxoSmithKline, Collegeville, United States of America
| | - Scott B. Berger
- Pattern Recognition Receptor Discovery Performance Unit, Immuno-Inflammation Therapeutic Area, GlaxoSmithKline, Collegeville, United States of America
| | - Joshi Ramanjulu
- Pattern Recognition Receptor Discovery Performance Unit, Immuno-Inflammation Therapeutic Area, GlaxoSmithKline, Collegeville, United States of America
| | - John G. Emery
- Pattern Recognition Receptor Discovery Performance Unit, Immuno-Inflammation Therapeutic Area, GlaxoSmithKline, Collegeville, United States of America
| | - Peter J. Gough
- Pattern Recognition Receptor Discovery Performance Unit, Immuno-Inflammation Therapeutic Area, GlaxoSmithKline, Collegeville, United States of America
| | - John Bertin
- Pattern Recognition Receptor Discovery Performance Unit, Immuno-Inflammation Therapeutic Area, GlaxoSmithKline, Collegeville, United States of America
- * E-mail:
| | - Kevin P. Foley
- Pattern Recognition Receptor Discovery Performance Unit, Immuno-Inflammation Therapeutic Area, GlaxoSmithKline, Collegeville, United States of America
| |
Collapse
|
38
|
Nunomura S, Ohtsubo-Yoshioka M, Okayama Y, Terui T, Ra C. FcRγpromotes contact hypersensitivity to oxazolone without affecting the contact sensitisation process in B6 mice. Exp Dermatol 2015; 24:204-8. [DOI: 10.1111/exd.12622] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/10/2014] [Indexed: 11/25/2022]
Affiliation(s)
- Satoshi Nunomura
- Department of Dermatology; Nihon University School of Medicine; Tokyo Japan
- Allergy and Immunology Group; Research Institute of Medical Science; Nihon University School of Medicine; Tokyo Japan
| | | | - Yoshimichi Okayama
- Allergy and Immunology Group; Research Institute of Medical Science; Nihon University School of Medicine; Tokyo Japan
| | - Tadashi Terui
- Department of Dermatology; Nihon University School of Medicine; Tokyo Japan
| | - Chisei Ra
- Allergy and Immunology Group; Research Institute of Medical Science; Nihon University School of Medicine; Tokyo Japan
- Asahi Hospital; Chiba Japan
- Department of Microbiology; Nihon University School of Medicine; Tokyo Japan
| |
Collapse
|
39
|
Danelli L, Frossi B, Gri G, Mion F, Guarnotta C, Bongiovanni L, Tripodo C, Mariuzzi L, Marzinotto S, Rigoni A, Blank U, Colombo MP, Pucillo CE. Mast cells boost myeloid-derived suppressor cell activity and contribute to the development of tumor-favoring microenvironment. Cancer Immunol Res 2014; 3:85-95. [PMID: 25351848 DOI: 10.1158/2326-6066.cir-14-0102] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Inflammation plays crucial roles at different stages of tumor development and may lead to the failure of immune surveillance and immunotherapy. Myeloid-derived suppressor cells (MDSC) are one of the major components of the immune-suppressive network that favors tumor growth, and their interaction with mast cells is emerging as critical for the outcome of the tumor-associated immune response. Herein, we showed the occurrence of cell-to-cell interactions between MDSCs and mast cells in the mucosa of patients with colon carcinoma and in the colon and spleen of tumor-bearing mice. Furthermore, we demonstrated that the CT-26 colon cancer cells induced the accumulation of CD11b(+)Gr1(+) immature MDSCs and the recruitment of protumoral mast cells at the tumor site. Using ex vivo analyses, we showed that mast cells have the ability to increase the suppressive properties of spleen-derived monocytic MDSCs, through a mechanism involving IFNγ and nitric oxide production. In addition, we demonstrated that the CD40:CD40L cross-talk between the two cell populations is responsible for the instauration of a proinflammatory microenvironment and for the increase in the production of mediators that can further support MDSC mobilization and tumor growth. In light of these results, interfering with the MDSC:mast cell axis could be a promising approach to abrogate MDSC-related immune suppression and to improve the antitumor immune response.
Collapse
Affiliation(s)
- Luca Danelli
- Department of Medical and Biological Science, University of Udine, Udine, Italy. Inserm UMRS-1149; CNRS ERL 8252; Université Paris Diderot, Sorbonne Paris Cite, Laboratoire d'excellence INFLAMEX, 75018 Paris, France
| | - Barbara Frossi
- Department of Medical and Biological Science, University of Udine, Udine, Italy
| | - Giorgia Gri
- Department of Medical and Biological Science, University of Udine, Udine, Italy
| | - Francesca Mion
- Department of Medical and Biological Science, University of Udine, Udine, Italy
| | - Carla Guarnotta
- Department of Health Science, University of Palermo, Palermo, Italy
| | | | - Claudio Tripodo
- Department of Health Science, University of Palermo, Palermo, Italy
| | - Laura Mariuzzi
- Department of Medical and Biological Science, University of Udine, Udine, Italy
| | - Stefania Marzinotto
- Department of Medical and Biological Science, University of Udine, Udine, Italy
| | - Alice Rigoni
- Department of Experimental Oncology and Molecular Medicine, Fondazione IRCCS Istituto Nazionale Tumori, Milan, Italy
| | - Ulrich Blank
- Inserm UMRS-1149; CNRS ERL 8252; Université Paris Diderot, Sorbonne Paris Cite, Laboratoire d'excellence INFLAMEX, 75018 Paris, France
| | - Mario P Colombo
- Department of Experimental Oncology and Molecular Medicine, Fondazione IRCCS Istituto Nazionale Tumori, Milan, Italy.
| | - Carlo E Pucillo
- Department of Medical and Biological Science, University of Udine, Udine, Italy.
| |
Collapse
|
40
|
Blank U, Madera-Salcedo IK, Danelli L, Claver J, Tiwari N, Sánchez-Miranda E, Vázquez-Victorio G, Ramírez-Valadez KA, Macias-Silva M, González-Espinosa C. Vesicular trafficking and signaling for cytokine and chemokine secretion in mast cells. Front Immunol 2014; 5:453. [PMID: 25295038 PMCID: PMC4170139 DOI: 10.3389/fimmu.2014.00453] [Citation(s) in RCA: 89] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2014] [Accepted: 09/05/2014] [Indexed: 12/31/2022] Open
Abstract
Upon activation mast cells (MCs) secrete numerous inflammatory compounds stored in their cytoplasmic secretory granules by a process called anaphylactic degranulation, which is responsible for type I hypersensitivity responses. Prestored mediators include histamine and MC proteases but also some cytokines and growth factors making them available within minutes for a maximal biological effect. Degranulation is followed by the de novo synthesis of lipid mediators such as prostaglandins and leukotrienes as well as a vast array of cytokines, chemokines, and growth factors, which are responsible for late phase inflammatory responses. While lipid mediators diffuse freely out of the cell through lipid bilayers, both anaphylactic degranulation and secretion of cytokines, chemokines, and growth factors depends on highly regulated vesicular trafficking steps that occur along the secretory pathway starting with the translocation of proteins to the endoplasmic reticulum. Vesicular trafficking in MCs also intersects with endocytic routes, notably to form specialized cytoplasmic granules called secretory lysosomes. Some of the mediators like histamine reach granules via specific vesicular monoamine transporters directly from the cytoplasm. In this review, we try to summarize the available data on granule biogenesis and signaling events that coordinate the complex steps that lead to the release of the inflammatory mediators from the various vesicular carriers in MCs.
Collapse
Affiliation(s)
- Ulrich Blank
- INSERM UMRS 1149 , Paris , France ; CNRS ERL8252 , Paris , France ; Université Paris Diderot, Sorbonne Paris Cité, Laboratoire d'excellence INFLAMEX , Paris , France
| | - Iris Karina Madera-Salcedo
- INSERM UMRS 1149 , Paris , France ; CNRS ERL8252 , Paris , France ; Université Paris Diderot, Sorbonne Paris Cité, Laboratoire d'excellence INFLAMEX , Paris , France
| | - Luca Danelli
- INSERM UMRS 1149 , Paris , France ; CNRS ERL8252 , Paris , France ; Université Paris Diderot, Sorbonne Paris Cité, Laboratoire d'excellence INFLAMEX , Paris , France
| | - Julien Claver
- INSERM UMRS 1149 , Paris , France ; CNRS ERL8252 , Paris , France ; Université Paris Diderot, Sorbonne Paris Cité, Laboratoire d'excellence INFLAMEX , Paris , France
| | - Neeraj Tiwari
- INSERM UMRS 1149 , Paris , France ; CNRS ERL8252 , Paris , France ; Université Paris Diderot, Sorbonne Paris Cité, Laboratoire d'excellence INFLAMEX , Paris , France
| | | | - Genaro Vázquez-Victorio
- Instituto de Fisiología Celular, Universidad Nacional Autónoma de México , México City , México
| | | | - Marina Macias-Silva
- Instituto de Fisiología Celular, Universidad Nacional Autónoma de México , México City , México
| | | |
Collapse
|
41
|
IκB kinase 2 is essential for IgE-induced mast cell de novo cytokine production but not for degranulation. Cell Rep 2014; 8:1300-7. [PMID: 25176657 DOI: 10.1016/j.celrep.2014.07.046] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2013] [Revised: 06/06/2014] [Accepted: 07/25/2014] [Indexed: 01/12/2023] Open
Abstract
The immunoglobulin E (IgE)-mediated mast cell (MC) response is central to the pathogenesis of type I allergy and asthma. IκB kinase 2 (IKK2) was reported to couple IgE-induced signals to MC degranulation by phosphorylating the SNARE protein SNAP23. We investigated MC responses in mice with MC-specific inactivation of IKK2 or NF-κB essential modulator (NEMO), or animals with MC-specific expression of a mutant, constitutively active IKK2. We show that the IgE-induced late-phase cytokine response is reduced in mice lacking IKK2 or NEMO in MCs. However, anaphylactic in vivo responses of these animals are not different from those of control mice, and in vitro IKK2-deficient MCs readily phosphorylate SNAP23 and degranulate similarly to control cells in response to allergen or calcium ionophore. Constitutive overactivation of the NF-κB pathway has only slight effects on allergen-triggered MC responses. Thus, IKK2 is dispensable for MC degranulation, and the important question how IgE-induced signals trigger MC vesicle fusion remains open.
Collapse
|
42
|
Reber LL, Frossard N. Targeting mast cells in inflammatory diseases. Pharmacol Ther 2014; 142:416-35. [PMID: 24486828 DOI: 10.1016/j.pharmthera.2014.01.004] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2014] [Accepted: 01/24/2014] [Indexed: 12/24/2022]
Abstract
Although mast cells have long been known to play a critical role in anaphylaxis and other allergic diseases, they also participate in some innate immune responses and may even have some protective functions. Data from the study of mast cell-deficient mice have facilitated our understanding of some of the molecular mechanisms driving mast cell functions during both innate and adaptive immune responses. This review presents an overview of the biology of mast cells and their potential involvement in various inflammatory diseases. We then discuss some of the current pharmacological approaches used to target mast cells and their products in several diseases associated with mast cell activation.
Collapse
Affiliation(s)
- Laurent L Reber
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA.
| | - Nelly Frossard
- Laboratoire d'Innovation Thérapeutique, UMR 7200 CNRS-Université de Strasbourg, Faculté de Pharmacie, France
| |
Collapse
|
43
|
Heger K, Fierens K, Vahl JC, Aszodi A, Peschke K, Schenten D, Hammad H, Beyaert R, Saur D, van Loo G, Roers A, Lambrecht BN, Kool M, Schmidt-Supprian M. A20-deficient mast cells exacerbate inflammatory responses in vivo. PLoS Biol 2014; 12:e1001762. [PMID: 24453940 PMCID: PMC3891641 DOI: 10.1371/journal.pbio.1001762] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2013] [Accepted: 11/26/2013] [Indexed: 12/13/2022] Open
Abstract
Mast cells, best known as effector cells in pathogenic immunoglobulin-mediated responses, can sense a variety of “danger” signals; if manipulated to enhance their resulting inflammatory responses, they also exacerbate inflammatory diseases such as arthritis and lung inflammation. Mast cells are implicated in the pathogenesis of inflammatory and autoimmune diseases. However, this notion based on studies in mast cell-deficient mice is controversial. We therefore established an in vivo model for hyperactive mast cells by specifically ablating the NF-κB negative feedback regulator A20. While A20 deficiency did not affect mast cell degranulation, it resulted in amplified pro-inflammatory responses downstream of IgE/FcεRI, TLRs, IL-1R, and IL-33R. As a consequence house dust mite- and IL-33-driven lung inflammation, late phase cutaneous anaphylaxis, and collagen-induced arthritis were aggravated, in contrast to experimental autoimmune encephalomyelitis and immediate anaphylaxis. Our results provide in vivo evidence that hyperactive mast cells can exacerbate inflammatory disorders and define diseases that might benefit from therapeutic intervention with mast cell function. Mast cells mediate allergic and anaphylactic immune reactions. They are also equipped with innate pattern recognition, cytokine, and alarmin receptors, which induce inflammatory responses. Correlative studies in human patients hinted at roles for mast cells in autoimmune and inflammatory diseases. However, studies using mast cell-deficient mice have yielded contradictory results in this context. In this study we determined that A20, the negative feedback regulator, restricts inflammation downstream of the mast cell antigen (allergen) receptor module, innate pattern recognition receptors, and the alarmin receptor IL-33R. By mast cell–specific ablation of A20 we established a mouse model for exaggerated inflammatory but normal anaphylactic mast cell signaling. With these mice we evaluated the impact of increased mast cell-mediated inflammation under experimental conditions aimed at mimicking several inflammatory human diseases. Our results demonstrated that the lack of A20 from mast cells exacerbated disease in mouse models for rheumatoid arthritis and innate forms of asthma, but did not impact disease progression in a mouse model for multiple sclerosis. Our data provide direct evidence that enhanced inflammatory mast cell responses can contribute to disease pathology and do so via sensing and amplifying local inflammatory reactions driven by “danger” stimuli and/or tissue damage that leads to the release of alarmins.
Collapse
MESH Headings
- Anaphylaxis/chemically induced
- Anaphylaxis/immunology
- Anaphylaxis/metabolism
- Anaphylaxis/pathology
- Animals
- Arthritis, Experimental/chemically induced
- Arthritis, Experimental/immunology
- Arthritis, Experimental/metabolism
- Arthritis, Experimental/pathology
- Collagen Type II/administration & dosage
- Cysteine Endopeptidases
- DNA-Binding Proteins/deficiency
- DNA-Binding Proteins/genetics
- DNA-Binding Proteins/immunology
- Dinitrophenols/administration & dosage
- Encephalomyelitis, Autoimmune, Experimental/chemically induced
- Encephalomyelitis, Autoimmune, Experimental/immunology
- Encephalomyelitis, Autoimmune, Experimental/metabolism
- Encephalomyelitis, Autoimmune, Experimental/pathology
- Gene Expression
- Immunoglobulin E/genetics
- Immunoglobulin E/immunology
- Interleukin-1 Receptor-Like 1 Protein
- Interleukin-33
- Interleukins/genetics
- Interleukins/immunology
- Intracellular Signaling Peptides and Proteins/deficiency
- Intracellular Signaling Peptides and Proteins/genetics
- Intracellular Signaling Peptides and Proteins/immunology
- Lung/immunology
- Lung/metabolism
- Lung/pathology
- Male
- Mast Cells/immunology
- Mast Cells/metabolism
- Mast Cells/pathology
- Mice
- Mice, Transgenic
- Myelin-Oligodendrocyte Glycoprotein/administration & dosage
- NF-kappa B/genetics
- NF-kappa B/immunology
- Peptide Fragments/administration & dosage
- Pneumonia/chemically induced
- Pneumonia/immunology
- Pneumonia/metabolism
- Pneumonia/pathology
- Pyroglyphidae/immunology
- Receptors, IgE/genetics
- Receptors, IgE/immunology
- Receptors, Interleukin/genetics
- Receptors, Interleukin/immunology
- Receptors, Interleukin-1/genetics
- Receptors, Interleukin-1/immunology
- Serum Albumin, Bovine/administration & dosage
- Toll-Like Receptors/genetics
- Toll-Like Receptors/immunology
- Tumor Necrosis Factor alpha-Induced Protein 3
- Ubiquitin-Protein Ligases/deficiency
- Ubiquitin-Protein Ligases/genetics
- Ubiquitin-Protein Ligases/immunology
Collapse
Affiliation(s)
- Klaus Heger
- Molecular Immunology and Signal Transduction, Max Planck Institute of Biochemistry, Martinsried, Germany
| | - Kaat Fierens
- Laboratory of Immunoregulation, Department of Pulmonary Medicine, University Hospital Ghent, Ghent, Belgium
- Department for Molecular Biomedical Research, Vlaams Instituut voor Biotechnologie, Ghent, Belgium
| | - J. Christoph Vahl
- Molecular Immunology and Signal Transduction, Max Planck Institute of Biochemistry, Martinsried, Germany
| | - Attila Aszodi
- Department of Surgery, Ludwig Maximilians Universität, Munich, Germany
| | - Katrin Peschke
- Institute for Immunology, Technische Universität Dresden, Dresden, Germany
| | - Dominik Schenten
- Department of Immunobiology, Yale University School of Medicine, New Haven, Connecticut, United States of America
| | - Hamida Hammad
- Laboratory of Immunoregulation, Department of Pulmonary Medicine, University Hospital Ghent, Ghent, Belgium
- Department for Molecular Biomedical Research, Vlaams Instituut voor Biotechnologie, Ghent, Belgium
| | - Rudi Beyaert
- Department for Molecular Biomedical Research, Vlaams Instituut voor Biotechnologie, Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Dieter Saur
- II. Medizinische Klinik, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
| | - Geert van Loo
- Department for Molecular Biomedical Research, Vlaams Instituut voor Biotechnologie, Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Axel Roers
- Institute for Immunology, Technische Universität Dresden, Dresden, Germany
| | - Bart N. Lambrecht
- Laboratory of Immunoregulation, Department of Pulmonary Medicine, University Hospital Ghent, Ghent, Belgium
- Department for Molecular Biomedical Research, Vlaams Instituut voor Biotechnologie, Ghent, Belgium
- Department of Pulmonary Medicine, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Mirjam Kool
- Laboratory of Immunoregulation, Department of Pulmonary Medicine, University Hospital Ghent, Ghent, Belgium
- Department for Molecular Biomedical Research, Vlaams Instituut voor Biotechnologie, Ghent, Belgium
- Department of Pulmonary Medicine, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Marc Schmidt-Supprian
- Molecular Immunology and Signal Transduction, Max Planck Institute of Biochemistry, Martinsried, Germany
- * E-mail:
| |
Collapse
|
44
|
Kim DY, Kang TB, Shim DW, Sun X, Han JW, Ji YE, Kim TJ, Koppula S, Lee KH. Emodin attenuates A23187-induced mast cell degranulation and tumor necrosis factor-α secretion through protein kinase C and IκB kinase 2 signaling. Eur J Pharmacol 2013; 723:501-6. [PMID: 24239713 DOI: 10.1016/j.ejphar.2013.09.066] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2013] [Revised: 07/17/2013] [Accepted: 09/24/2013] [Indexed: 12/31/2022]
Abstract
Mast cells are known to play a pivotal role in allergic diseases. Cross-linking of the high-affinity IgE receptor (FcεRI) is known to be one of the major causes that lead to degranulation and allergic inflammation. An increase in intracellular calcium (Ca(2+)) concentration also triggers degranulation, bypassing receptor activation. Emodin (1,3,8-trihydroxy-6-methylanthraquinone) is known to exhibit a variety of pharmacological activities including anti-allergic effects. However, the detailed molecular mechanisms involved in exhibiting anti-allergic effects by emodin were remained to be clarified. In the present investigation we report the regulatory function of emodin on the allergic signal mediators through Ca(2+) ionophore activation in mast cells. Emodin significantly inhibited A23187-induced tumor necrosis factor-α production and degranulation through the attenuation of protein kinase C, IκB kinase 2, and soluble N-ethylmaleimide-sensitive fusion factor attachment protein receptor complex formation, bypassing FcεRI activation. Data from our study indicated that emodin acts by regulating multiple signaling pathways in inhibiting the allergic reactions in mast cells.
Collapse
Affiliation(s)
- Dong-Young Kim
- Department of Biotechnology, College of Biomedical & Health Science, Research Institute of Inflammatory Diseases, Konkuk University, Chungju, Chungbuk 380-701, Republic of Korea
| | - Tae-Bong Kang
- Department of Biotechnology, College of Biomedical & Health Science, Research Institute of Inflammatory Diseases, Konkuk University, Chungju, Chungbuk 380-701, Republic of Korea
| | - Do-Wan Shim
- Department of Biotechnology, College of Biomedical & Health Science, Research Institute of Inflammatory Diseases, Konkuk University, Chungju, Chungbuk 380-701, Republic of Korea
| | - Xiao Sun
- Department of Biotechnology, College of Biomedical & Health Science, Research Institute of Inflammatory Diseases, Konkuk University, Chungju, Chungbuk 380-701, Republic of Korea
| | - Ji-Won Han
- Department of Biotechnology, College of Biomedical & Health Science, Research Institute of Inflammatory Diseases, Konkuk University, Chungju, Chungbuk 380-701, Republic of Korea
| | - Young-Eun Ji
- Department of Biotechnology, College of Biomedical & Health Science, Research Institute of Inflammatory Diseases, Konkuk University, Chungju, Chungbuk 380-701, Republic of Korea
| | - Tack-Joong Kim
- Division of Biological Science and Technology, Yonsei University, Wonju, Republic of Korea
| | - Sushruta Koppula
- Department of Biotechnology, College of Biomedical & Health Science, Research Institute of Inflammatory Diseases, Konkuk University, Chungju, Chungbuk 380-701, Republic of Korea
| | - Kwang-Ho Lee
- Department of Biotechnology, College of Biomedical & Health Science, Research Institute of Inflammatory Diseases, Konkuk University, Chungju, Chungbuk 380-701, Republic of Korea.
| |
Collapse
|
45
|
Hosokawa J, Suzuki K, Nakagomi D, Tamachi T, Takatori H, Suto A, Nakajima H. Role of calcium ionophore A23187-induced activation of IkappaB kinase 2 in mast cells. Int Arch Allergy Immunol 2013; 161 Suppl 2:37-43. [PMID: 23711852 DOI: 10.1159/000350357] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Mast cells are known to play a pivotal role in allergic diseases by releasing granules containing histamine and other preformed chemical mediators. Cross-linking of high-affinity receptors for IgE (FcεRI) on mast cells results in rapid increases in intracellular free calcium concentration [Ca(2+)]i and consequent activation of many transcription factors, including NFAT, NF-κB, JNK and CREB. Ca(2+) signaling is essential for many cellular activities such as proliferation, gene expression and degranulation in mast cells. In addition to Ca(2+) signaling, previous reports have shown that IkappaB kinase 2 (IKK2 or IKKβ), a central component of the IKK complex mediating NF-κB activation, also plays a crucial role in FcεRI-mediated degranulation and cytokine production. Moreover, it has been demonstrated that activation of PKCβ, a calcium-dependent PKC isoform, leads to IKK2 activation in many cell types. However, the roles of Ca(2+) signaling and PKCβ in the activation of IKK2 in mast cells remain largely unknown. METHODS We investigated the effect of PKC inhibitor Gö6976 on calcium ionophore A23187-induced activation of IKK2 in mast cells. We also examined the role of IKK2 in A23187-induced NF-κB-dependent gene induction, degranulation, proinflammatory cytokine production and extracellular signal-regulated kinases 1 and 2 (ERK1/2) activation by using IKK2-deficient (IKK2(-/-)) fetal liver-derived mast cells (FLMCs). RESULTS A23187 activated IKK2 and NF-κB even in the presence of Gö6976 in mast cells. A23187-induced degranulation, cytokine production and activation of ERK1/2 were diminished in IKK2(-/-) FLMCs compared to those in wild-type FLMCs. CONCLUSIONS Ca(2+)-IKK2 signaling is involved in the degranulation and cytokine production in activated mast cells by a mechanism independent of PKCβ.
Collapse
Affiliation(s)
- Junichi Hosokawa
- Department of Allergy and Clinical Immunology, Graduate School of Medicine, Chiba University, Chiba, Japan
| | | | | | | | | | | | | |
Collapse
|
46
|
Pan YF, Dong LW, Wang M, Yang GZ, Zhang J, Li SX, Zhang B, Yang C, Li Z, Tan YX, Wang HY. Signal regulatory protein α negatively regulates mast-cell activation following FcεRI aggregation. Eur J Immunol 2013; 43:1598-607. [DOI: 10.1002/eji.201243031] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2012] [Revised: 01/31/2013] [Accepted: 03/08/2013] [Indexed: 12/13/2022]
Affiliation(s)
- Yu-fei Pan
- International Cooperation Laboratory on Signal Transduction; Eastern Hepatobiliary Surgery Institute, the Second Military Medical University; Shanghai; P. R. China
| | - Li-wei Dong
- International Cooperation Laboratory on Signal Transduction; Eastern Hepatobiliary Surgery Institute, the Second Military Medical University; Shanghai; P. R. China
| | - Min Wang
- International Cooperation Laboratory on Signal Transduction; Eastern Hepatobiliary Surgery Institute, the Second Military Medical University; Shanghai; P. R. China
| | - Guang-zhen Yang
- International Cooperation Laboratory on Signal Transduction; Eastern Hepatobiliary Surgery Institute, the Second Military Medical University; Shanghai; P. R. China
| | - Jian Zhang
- International Cooperation Laboratory on Signal Transduction; Eastern Hepatobiliary Surgery Institute, the Second Military Medical University; Shanghai; P. R. China
| | - Shuang-xi Li
- International Cooperation Laboratory on Signal Transduction; Eastern Hepatobiliary Surgery Institute, the Second Military Medical University; Shanghai; P. R. China
| | - Bo Zhang
- International Cooperation Laboratory on Signal Transduction; Eastern Hepatobiliary Surgery Institute, the Second Military Medical University; Shanghai; P. R. China
| | - Chun Yang
- International Cooperation Laboratory on Signal Transduction; Eastern Hepatobiliary Surgery Institute, the Second Military Medical University; Shanghai; P. R. China
| | - Zhong Li
- International Cooperation Laboratory on Signal Transduction; Eastern Hepatobiliary Surgery Institute, the Second Military Medical University; Shanghai; P. R. China
| | - Ye-xiong Tan
- International Cooperation Laboratory on Signal Transduction; Eastern Hepatobiliary Surgery Institute, the Second Military Medical University; Shanghai; P. R. China
| | | |
Collapse
|
47
|
Graham AC, Hilmer KM, Zickovich JM, Obar JJ. Inflammatory response of mast cells during influenza A virus infection is mediated by active infection and RIG-I signaling. THE JOURNAL OF IMMUNOLOGY 2013; 190:4676-84. [PMID: 23526820 DOI: 10.4049/jimmunol.1202096] [Citation(s) in RCA: 72] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Influenza A virus (IAV) is a major respiratory pathogen of both humans and animals. The lung is protected from pathogens by alveolar epithelial cells, tissue-resident alveolar macrophages, dendritic cells, and mast cells. The role of alveolar epithelial cells, endothelial cells, and alveolar macrophages during IAV infection has been studied previously. In this study, we address the role of mast cells during IAV infection. Respiratory infection with A/WSN/33 causes significant disease and immunopathology in C57BL/6 mice but not in B6.Cg-Kit(W-sh) mice, which lack mast cells. During in vitro coculture, A/WSN/33 caused mast cells to release histamine, secrete cytokines and chemokines, and produce leukotrienes. Moreover, when mast cells were infected with IAV, the virus did not replicate within mast cells. Importantly, human H1N1, H3N2, and influenza B virus isolates also could activate mast cells in vitro. Mast cell production of cytokines and chemokines occurs in a RIG-I/MAVS-dependent mechanism; in contrast, histamine production occurred through a RIG-I/MAVS-independent mechanism. Our data highlight that, following IAV infection, the response of mast cells is controlled by multiple receptors. In conclusion, we identified a unique inflammatory cascade activated during IAV infection that could potentially be targeted to limit morbidity following IAV infection.
Collapse
Affiliation(s)
- Amy C Graham
- Department of Immunology and Infectious Diseases, Montana State University, Bozeman, MT 59718, USA
| | | | | | | |
Collapse
|
48
|
Marion S, Mazzolini J, Herit F, Bourdoncle P, Kambou-Pene N, Hailfinger S, Sachse M, Ruland J, Benmerah A, Echard A, Thome M, Niedergang F. The NF-κB signaling protein Bcl10 regulates actin dynamics by controlling AP1 and OCRL-bearing vesicles. Dev Cell 2013; 23:954-67. [PMID: 23153494 DOI: 10.1016/j.devcel.2012.09.021] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2012] [Revised: 08/18/2012] [Accepted: 09/21/2012] [Indexed: 11/30/2022]
Abstract
The protein Bcl10 contributes to adaptive and innate immunity through the assembly of a signaling complex that plays a key role in antigen receptor and FcR-induced NF-κB activation. Here we demonstrate that Bcl10 has an NF-κB-independent role in actin and membrane remodeling downstream of FcR in human macrophages. Depletion of Bcl10 impaired Rac1 and PI3K activation and led to an abortive phagocytic cup rich in PI(4,5)P(2), Cdc42, and F-actin, which could be rescued with low doses of F-actin depolymerizing drugs. Unexpectedly, we found Bcl10 in a complex with the clathrin adaptors AP1 and EpsinR. In particular, Bcl10 was required to locally deliver the vesicular OCRL phosphatase that regulates PI(4,5)P(2) and F-actin turnover, both crucial for the completion of phagosome closure. Thus, we identify Bcl10 as an early coordinator of NF-κB-mediated immune response with endosomal trafficking and signaling to F-actin remodeling.
Collapse
Affiliation(s)
- Sabrina Marion
- Inserm, U1016, Institut Cochin, Paris, France; CNRS, UMR 8104, Paris, France
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Shin J, Zhang P, Wang S, Wu J, Guan Z, Zhong XP. Negative control of mast cell degranulation and the anaphylactic response by the phosphatase lipin1. Eur J Immunol 2012; 43:240-8. [PMID: 23065777 DOI: 10.1002/eji.201242571] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2012] [Revised: 09/14/2012] [Accepted: 10/10/2012] [Indexed: 11/11/2022]
Abstract
Mast cells play a critical role in the pathogenesis of allergic diseases; however, how mast cell function is regulated is still not well understood. Both phosphatidic acid (PA) and diacylglycerol (DAG) are important secondary messengers involved in mast cell activ-ation. Lipin1 is a phosphatidate phosphatase that hydrolyzes PA to produce DAG, but the role of lipin1 in mast cell function has been thus far unknown. Here we show that lipin1 is an important and selective inhibitor of mast cell degranulation. Lipin1 deficiency enhanced FcεRI-mediated β-hexosaminidase and prostaglandin D2 release from mast cells in vitro and exacerbated the passive systemic anaphylaxis reaction in vivo. Lipin1 deficiency, however, did not exert obvious effects on IL-6 or TNF-α production following FcεRI engagement. FcεRI-induced PKC and SNAP-23 phosphorylation were augmented in the lipin1-deficient mast cells. Moreover, inhibition of PKC activity reduced SNAP-23 phosphorylation and mast cell degranulation in lipin1-deficient mast cells. Together, our findings suggest that lipin1 may negatively control mast cell degranulation and the anaphylactic response through inhibiting the PKC-SNAP-23 pathway.
Collapse
Affiliation(s)
- Jinwook Shin
- Division of Allergy and Immunology, Department of Pediatrics, Duke University Medical Center, Durham, NC 27710, USA
| | | | | | | | | | | |
Collapse
|
50
|
Price MM, Oskeritzian CA, Falanga YT, Harikumar KB, Allegood JC, Alvarez SE, Conrad D, Ryan JJ, Milstien S, Spiegel S. A specific sphingosine kinase 1 inhibitor attenuates airway hyperresponsiveness and inflammation in a mast cell-dependent murine model of allergic asthma. J Allergy Clin Immunol 2012; 131:501-11.e1. [PMID: 22939756 DOI: 10.1016/j.jaci.2012.07.014] [Citation(s) in RCA: 128] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2011] [Revised: 06/15/2012] [Accepted: 07/10/2012] [Indexed: 10/28/2022]
Abstract
BACKGROUND Sphingosine-1-phosphate (S1P), which is produced by 2 sphingosine kinase (SphK) isoenzymes, SphK1 and SphK2, has been implicated in IgE-mediated mast cell responses. However, studies of allergic inflammation in isotype-specific SphK knockout mice have not clarified their contribution, and the role that S1P plays in vivo in a mast cell- and IgE-dependent murine model of allergic asthma has not yet been examined. OBJECTIVE We used an isoenzyme-specific SphK1 inhibitor, SK1-I, to investigate the contributions of S1P and SphK1 to mast cell-dependent airway hyperresponsiveness (AHR) and airway inflammation in mice. METHODS Allergic airway inflammation and AHR were examined in a mast cell-dependent murine model of ovalbumin (OVA)-induced asthma. C57BL/6 mice received intranasal delivery of SK1-I before sensitization and challenge with OVA or only before challenge. RESULTS SK1-I inhibited antigen-dependent activation of human and murine mast cells and suppressed activation of nuclear factor κB (NF-κB), a master transcription factor that regulates the expression of proinflammatory cytokines. SK1-I treatment of mice sensitized to OVA in the absence of adjuvant, in which mast cell-dependent allergic inflammation develops, significantly reduced OVA-induced AHR to methacholine; decreased numbers of eosinophils and levels of the cytokines IL-4, IL-5, IL-6, IL-13, IFN-γ, and TNF-α and the chemokines eotaxin and CCL2 in bronchoalveolar lavage fluid; and decreased pulmonary inflammation, as well as activation of NF-κB in the lungs. CONCLUSION S1P and SphK1 play important roles in mast cell-dependent, OVA-induced allergic inflammation and AHR, in part by regulating the NF-κB pathway.
Collapse
Affiliation(s)
- Megan M Price
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth University School of Medicine, Richmond, Va 23298-0614, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|