1
|
Brown PA. Transcriptomic signatures of atheroresistance in the human atrium and ventricle highlight potential candidates for targeted atherosclerosis therapeutics. Biochem Biophys Rep 2025; 42:102007. [PMID: 40248137 PMCID: PMC12004712 DOI: 10.1016/j.bbrep.2025.102007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2024] [Revised: 03/28/2025] [Accepted: 04/04/2025] [Indexed: 04/19/2025] Open
Abstract
Atherosclerosis risk is not uniform throughout the cardiovascular system. This study therefore aimed to compare the transcriptomes of atheroresistant human atrium and ventricle with atheroprone coronary arteries to identify transcriptomic signatures of atheroresistance and potential targets for atherosclerosis therapeutics. Using publicly available gene read counts, differentially expressed genes between the atrium, ventricle, and coronary artery were identified for each contrast and validated against the Swiss Institute of Bioinformatics' Bgee database. Over-representation analysis and active-subnetwork-oriented enrichment assessment then identified enriched terms, which were grouped into endothelial dysfunction-related processes. Potential biological significance was further explored with pathway analysis. Among 21474 features, 12656 differentially expressed genes were identified across the three contrasts and associated with 1215 enriched terms. There were 315 down-regulated and 133 up-regulated genes associated with endothelial dysfunction-related processes across the contrasts, including immune modulators, cell adhesion molecules, and lipid metabolism- and coagulation-related molecules. Differentially expressed genes were associated with six down-regulated Kyoto Encyclopedia of Genes and Genomes pathways, related to immune cell and associated endothelium functions. Review of regulated genes associated with endothelial dysfunction-related processes and included in these pathways, indicate immune cell-associated B cell scaffold protein with ankyrin repeats 1, as well as arterial endothelial cell-associated vascular cell adhesion molecule 1 and cadherin 5, as potential atherosclerosis targets.
Collapse
Affiliation(s)
- Paul A. Brown
- Department of Basic Medical Sciences, Faculty of Medical Sciences Teaching and Research Complex, The University of the West Indies, Mona, Kingston 7, Jamaica
| |
Collapse
|
2
|
Sim KH, Park KM, Lee C, Lee YJ. Perfluorooctane sulfonate attenuates IgE/Ag-stimulated mast cell activation and anaphylactic responses via activating SHP-1 pathway. CHEMOSPHERE 2025; 372:144119. [PMID: 39827624 DOI: 10.1016/j.chemosphere.2025.144119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/29/2024] [Revised: 11/26/2024] [Accepted: 01/12/2025] [Indexed: 01/22/2025]
Abstract
Perfluorooctane sulfonate (PFOS), a widely distributed and persistent organic pollutant, is known to cause immune dysfunction. In a previous study, we reported that PFOS modestly increases mast cell activation. However, its effects on FcεRI (a high-affinity IgE receptor)-mediated mast cell activation, a pivotal process in inflammatory allergic reactions and innate immunity, have not been clearly demonstrated. In this study, we investigated the effects of PFOS on IgE/Ag (antigen)-stimulated mast cell activation and the underlying mechanisms using bone marrow-derived mast cells (BMMCs) and a passive cutaneous anaphylaxis (PCA) mouse model. Oral administration of PFOS attenuated IgE/Ag-stimulated PCA responses. In the BMMCs model, PFOS reduced IgE/Ag-stimulated degranulation, intracellular Ca2+ levels, eicosanoid synthesis, and mRNA expression of pro-inflammatory cytokines. Consistently, PFOS decreased the phosphorylation of Syk and Lck, central tyrosine kinases in IgE/Ag-stimulated mast cell activation, along with their downstream signaling molecules (PLCγ1, AKT, and MAPKs), through the activation of tyrosine phosphatase Src homology region 2 domain-containing phosphatase-1. Additionally, PFOS reduced the phosphorylation of FcεRI-associated tyrosine kinases Fyn and Lyn. Fluorine-19 nuclear magnetic resonance spectroscopy revealed reduced fluorine signals of PFOS upon interaction with the plasma membrane, suggesting that PFOS accumulates in plasma membranes and interferes with FcεRI signaling by acting upstream, close to the membrane. Moreover, PFOS attenuated lipopolysaccharide-stimulated mRNA expression of TNFα and IL-6. In conclusion, PFOS exposure disrupts FcεRI-mediated allergic responses and modulates innate immune responses.
Collapse
Affiliation(s)
- Kyeong Hwa Sim
- Department of Pharmacology/Toxicology, School of Medicine, Daegu Catholic University, Daegu, Republic of Korea
| | - Kyeng Min Park
- Department of Biochemistry, School of Medicine, Daegu Catholic University, Daegu, Republic of Korea
| | - Chuhee Lee
- Department of Biochemistry & Molecular Biology, School of Medicine, Yeungnam University, Daegu, Republic of Korea
| | - Youn Ju Lee
- Department of Pharmacology/Toxicology, School of Medicine, Daegu Catholic University, Daegu, Republic of Korea.
| |
Collapse
|
3
|
Sim KH, Lee E, Shrestha P, Choi BH, Hong J, Lee YJ. Isobavachin attenuates FcεRI-mediated inflammatory allergic responses by regulating SHP-1-dependent Fyn/Lyn/Syk/Lck signaling. Biochem Pharmacol 2025; 232:116698. [PMID: 39643121 DOI: 10.1016/j.bcp.2024.116698] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2024] [Revised: 11/30/2024] [Accepted: 12/03/2024] [Indexed: 12/09/2024]
Abstract
Isobavachin, isolated from Psoralea corylifolia L. exhibits therapeutic potential for osteoporosis or skin disease. Here, we evaluated the pharmacological effects of isobavachin on IgE-dependent inflammatory allergic reactions, as well as the underlying mechanisms, in bone marrow-derived mast cells and a mouse model of passive cutaneous anaphylaxis (PCA). Isobavachin reduced IgE/Ag-stimulated degranulation, eicosanoid (leukotriene C4 and prostaglandin D2) generation, and release of pro-inflammatory cytokines (tumor necrosis factor-α (TNF-α) and interleukin (IL)-6). Mechanistic studies revealed that isobavachin suppressed activation of Fyn, Lyn, spleen tyrosine kinase (Syk), and lymphocyte-specific-protein-kinase (Lck), receptor-proximal tyrosine kinases that initiate and play a central role in FcɛRI-mediated mast cell activation, as well as their common downstream signaling molecules including linker for activation of T cells, phospholipase Cγ1, AKT, mitogen-activated protein kinases (MAPKs), and intracellular Ca2+. Additionally, isobavachin increased phosphorylation of Src homology region 2 domain-containing phosphatase-1 (SHP-1), thereby strengthening its interaction with Syk and Lck as well as Fyn and Lyn, resulting in de-phosphorylation of these proximal tyrosine kinases. Genetic knockdown of SHP-1 reversed the inhibitory effects of isobavachin on mast cell activation, as well as the related signaling pathways, indicating that the inhibitory effects of isobavachin are mediated by negative regulation of SHP-1-dependent Fyn, Lyn, Syk and Lck. The anti-inflammatory properties of isobavachin were also examined in macrophages. Isobavachin suppressed production of lipopolysaccharide-stimulated production of pro-inflammatory cytokines and nitric oxide. Furthermore, oral administration of isobavachin attenuated mast cell-mediated PCA reactions in mice. These results suggest that isobavachin is a potential treatment for mast cell-mediated allergic inflammatory diseases.
Collapse
Affiliation(s)
- Kyeong Hwa Sim
- Department of Pharmacology, School of Medicine, Daegu Catholic University, 33 Duryugongwon-ro 17-gil, Nam-gu, Daegu 42472, Republic of Korea
| | - Eunkyung Lee
- Department of Korean Medicine Development, National Institute for Korean Medicine Development, Gyeongsan 38540, Republic of Korea
| | - Prafulla Shrestha
- Department of Pharmacology, School of Medicine, Daegu Catholic University, 33 Duryugongwon-ro 17-gil, Nam-gu, Daegu 42472, Republic of Korea
| | - Bo-Hyun Choi
- Department of Pharmacology, School of Medicine, Daegu Catholic University, 33 Duryugongwon-ro 17-gil, Nam-gu, Daegu 42472, Republic of Korea
| | - Jaewoo Hong
- Department of Physiology, School of Medicine, Daegu Catholic University, 33 Duryugongwon-ro 17-gil, Nam-gu, Daegu 42472, Republic of Korea; Department of Companion Animal Health, Daegu Catholic University, Gyeongsan, Gyeongbuk 38430, Republic of Korea; Eversummer Lab, Daegu Catholic University, Gyeongsan, Gyeongbuk 38430, Republic of Korea; Department of Research and Development, CaniCatiCare Inc., Daegu 42078, Republic of Korea
| | - Youn Ju Lee
- Department of Pharmacology, School of Medicine, Daegu Catholic University, 33 Duryugongwon-ro 17-gil, Nam-gu, Daegu 42472, Republic of Korea.
| |
Collapse
|
4
|
Hao G, Han S, Xiao Z, Shen J, Zhao Y, Hao Q. Synovial mast cells and osteoarthritis: Current understandings and future perspectives. Heliyon 2024; 10:e41003. [PMID: 39720069 PMCID: PMC11665477 DOI: 10.1016/j.heliyon.2024.e41003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Revised: 10/29/2024] [Accepted: 12/04/2024] [Indexed: 12/26/2024] Open
Abstract
Osteoarthritis (OA) is a prevalent joint disease worldwide that significantly impacts the quality of life of individuals, particularly those in middle-aged and elderly populations. OA was initially considered as non-inflammatory arthritis, but recent studies have identified a substantial number of immune responses in OA, leading to the recognition of inflammation as a key factor in its pathogenesis. An increasing number of studies have found that mast cell (MC) and MC-secreted inflammatory mediators and cytokines are notably increased in the synovial fluid of OA patients, indicating a potential association between MCs and the onset and progression of synovial inflammation. The present review aims to summarize the significance and mechanism of MCs in the pathogenesis of OA. Meanwhile, we also discuss the clinical potential of using MCs as therapeutic target for OA therapy. Modulating the activities of MCs or the mediators of MCs in the synovial fluid inflammatory microenvironment will be promising new options for the treatment of OA.
Collapse
Affiliation(s)
- Guanghui Hao
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China
- Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Southwest Medical University, Luzhou, Sichuan, China
- South Sichuan Institute of Translational Medicine, Luzhou, Sichuan, China
| | - Shanqian Han
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China
- Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Southwest Medical University, Luzhou, Sichuan, China
- South Sichuan Institute of Translational Medicine, Luzhou, Sichuan, China
| | - Zhangang Xiao
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China
- Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Southwest Medical University, Luzhou, Sichuan, China
- South Sichuan Institute of Translational Medicine, Luzhou, Sichuan, China
| | - Jing Shen
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China
- Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Southwest Medical University, Luzhou, Sichuan, China
- South Sichuan Institute of Translational Medicine, Luzhou, Sichuan, China
| | - Yueshui Zhao
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China
- Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Southwest Medical University, Luzhou, Sichuan, China
- South Sichuan Institute of Translational Medicine, Luzhou, Sichuan, China
| | - Qi Hao
- Department of Joint Surgery, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, Sichuan, China
- The Third People's Hospital of Longmatan District, Luzhou, Sichuan, China
| |
Collapse
|
5
|
Dangelmaier C, Vari HR, Vajipayajula DN, Elzoheiry M, Wright M, Iyer A, Tsygankov AY, Kunapuli SP. Phosphorylation of (Ser 291) in the linker insert of Syk negatively regulates ITAM signaling in platelets. Platelets 2024; 35:2369766. [PMID: 38904212 PMCID: PMC11322839 DOI: 10.1080/09537104.2024.2369766] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/22/2024]
Abstract
Receptor-induced tyrosine phosphorylation of spleen tyrosine kinase (Syk) has been studied extensively in hematopoietic cells. Metabolic mapping and high-resolution mass spectrometry, however, indicate that one of the most frequently detected phosphorylation sites encompassed S297 (S291 in mice) located within the linker B region of Syk. It has been reported that Protein kinase C (PKC) phosphorylates Syk S297, thus influencing Syk activity. However, conflicting studies suggest that this phosphorylation enhances as well as reduces Syk activity. To clarify the function of this site, we generated Syk S291A knock-in mice. We used platelets as a model system as they possess Glycoprotein VI (GPVI), a receptor containing an immunoreceptor tyrosine-based activation motif (ITAM) which transduces signals through Syk. Our analysis of the homozygous mice indicated that the knock-in platelets express only one isoform of Syk, while the wild-type expresses two isoforms at 69 and 66 kDa. When the GPVI receptor was activated with collagen-related peptide (CRP), we observed an increase in functional responses and phosphorylations in Syk S291A platelets. This potentiation did not occur with AYPGKF or 2-MeSADP, although they also activate PKC isoforms. Although there was potentiation of platelet functional responses, there was no difference in tail bleeding times. However, the time to occlusion in the FeCl3 injury model was enhanced. These data indicate that the effects of Syk S291 phosphorylation represent a significant outcome on platelet activation and signaling in vitro but also reveals its multifaceted nature demonstrated by the differential effects on physiological responses in vivo.
Collapse
Affiliation(s)
- Carol Dangelmaier
- Sol Sherry Thrombosis Research Center, Department of Cardiovascular Sciences, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA
| | - Hymavathi Reddy Vari
- Sol Sherry Thrombosis Research Center, Department of Cardiovascular Sciences, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA
| | - Dhruv N Vajipayajula
- Sol Sherry Thrombosis Research Center, Department of Cardiovascular Sciences, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA
| | - Manal Elzoheiry
- Sol Sherry Thrombosis Research Center, Department of Cardiovascular Sciences, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA
| | - Monica Wright
- Sol Sherry Thrombosis Research Center, Department of Cardiovascular Sciences, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA
| | - Ashvin Iyer
- Sol Sherry Thrombosis Research Center, Department of Cardiovascular Sciences, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA
| | - Alexander Y Tsygankov
- Sol Sherry Thrombosis Research Center, Department of Cardiovascular Sciences, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA
| | - Satya P Kunapuli
- Sol Sherry Thrombosis Research Center, Department of Cardiovascular Sciences, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA
| |
Collapse
|
6
|
Obeng B, Bennett LJ, West BE, Wagner DJ, Fleming PJ, Tasker MN, Lorenger MK, Smith DR, Systuk T, Plummer SM, Eom J, Paine MD, Frangos CT, Wilczek MP, Shim JK, Maginnis MS, Gosse JA. Anti-microbial cetylpyridinium chloride suppresses mast cell function by targeting tyrosine phosphorylation of Syk kinase. J Immunotoxicol 2024; 21:2443397. [PMID: 39815634 PMCID: PMC11827644 DOI: 10.1080/1547691x.2024.2443397] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Revised: 11/22/2024] [Accepted: 12/12/2024] [Indexed: 01/18/2025] Open
Abstract
Cetylpyridinium chloride (CPC) is a quaternary ammonium antimicrobial used in numerous personal care products, human food, cosmetic products, and cleaning solutions. Yet, there is minimal published data on CPC effects on eukaryotes, immune signaling, and human health. Previously, it was shown that low-micromolar CPC inhibits rat mast cell function by inhibiting antigen (Ag)-stimulated Ca2+ mobilization, microtubule polymerization, and degranulation. In the current study, these findings are extended to human mast cells (LAD2); this paper presents data indicating that a mechanism of action for CPC might center on its positively-charged quaternary nitrogen in its pyridinium headgroup. The inhibitory effect of CPC was independent of signaling platform receptor architecture. Tyrosine phosphorylation events are a trigger of Ca2+ mobilization necessary for degranulation. CPC inhibits global tyrosine phosphorylation in Ag-stimulated mast cells. Specifically, CPC inhibits tyrosine phosphorylation of specific key players Syk kinase and LAT, a substrate of Syk. In contrast, CPC did not affect Lyn kinase phosphorylation. Thus, a root mechanism for CPC effect might be electrostatic disruption of particular tyrosine phosphorylation events essential for signaling. This work presented here outlines biochemical mechanisms underlying the effects of CPC on immune signaling.
Collapse
Affiliation(s)
- Bright Obeng
- Department of Molecular and Biomedical Sciences, University of Maine, Orono, ME
| | - Lucas J. Bennett
- Department of Molecular and Biomedical Sciences, University of Maine, Orono, ME
| | - Bailey E. West
- Department of Molecular and Biomedical Sciences, University of Maine, Orono, ME
| | - Dylan J. Wagner
- Department of Molecular and Biomedical Sciences, University of Maine, Orono, ME
| | - Patrick J. Fleming
- Department of Molecular and Biomedical Sciences, University of Maine, Orono, ME
| | - Morgan N. Tasker
- Department of Molecular and Biomedical Sciences, University of Maine, Orono, ME
| | | | - Dorothy R. Smith
- Department of Molecular and Biomedical Sciences, University of Maine, Orono, ME
| | - Tetiana Systuk
- Department of Molecular and Biomedical Sciences, University of Maine, Orono, ME
| | - Sydni M. Plummer
- Department of Molecular and Biomedical Sciences, University of Maine, Orono, ME
| | - Jeongwon Eom
- Department of Molecular and Biomedical Sciences, University of Maine, Orono, ME
| | - Marissa D. Paine
- Department of Molecular and Biomedical Sciences, University of Maine, Orono, ME
| | | | - Michael P. Wilczek
- Department of Molecular and Biomedical Sciences, University of Maine, Orono, ME
| | - Juyoung K. Shim
- Department of Biology, University of Maine Augusta, Augusta, ME
| | - Melissa S. Maginnis
- Department of Molecular and Biomedical Sciences, University of Maine, Orono, ME
| | - Julie A. Gosse
- Department of Molecular and Biomedical Sciences, University of Maine, Orono, ME
| |
Collapse
|
7
|
Choi MY, Jo MG, Min KY, Kim B, Kim Y, Choi WS. Antimicrobial Peptide Pro10-1D Exhibits Anti-Allergic Activity: A Promising Therapeutic Candidate. Int J Mol Sci 2024; 25:12138. [PMID: 39596204 PMCID: PMC11594534 DOI: 10.3390/ijms252212138] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Revised: 11/08/2024] [Accepted: 11/11/2024] [Indexed: 11/28/2024] Open
Abstract
Although antimicrobial peptides (AMPs) exhibit a range of biological functions, reports on AMPs with therapeutic effects in allergic disorders are limited. In this study, we investigated the anti-allergic effects of Pro10-1D, a 10-meric AMP derived from insect defensin protaetiamycine. Our findings demonstrate that Pro10-1D effectively inhibits antigen-induced degranulation of mast cells (MCs) with IC50 values of approximately 11.6 μM for RBL-2H3 cells and 2.7 μM for bone marrow-derived MCs. Furthermore, Pro10-1D suppressed the secretion of cytokines with IC50 values of approximately 2.8 μM for IL-4 and approximately 8.6 μM for TNF-α. Mechanistically, Pro10-1D inhibited the Syk-LAT-PLCγ1 signaling pathway in MCs and decreased the activation of mitogen-activated protein kinases (MAPKs). Pro10-1D demonstrated a dose-dependent reduction in IgE-mediated passive cutaneous anaphylaxis in mice with an ED50 value of approximately 7.6 mg/kg. Further investigation revealed that Pro10-1D significantly reduced the activity of key kinases Fyn and Lyn, which are critical in the initial phase of the FcεRI-mediated signaling pathway, with IC50 values of approximately 22.6 μM for Fyn and approximately 1.5 μM for Lyn. Collectively, these findings suggest that Pro10-1D represents a novel therapeutic candidate for the treatment of IgE-mediated allergic disorders by targeting the Lyn/Fyn Src family kinases in MCs.
Collapse
Affiliation(s)
- Min Yeong Choi
- Department of Immunology, College of Medicine, Konkuk University, Chungju 27478, Republic of Korea
| | - Min Geun Jo
- 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
| | - Byeongkwon Kim
- Department of Bioscience and Biotechnology, Konkuk University, Seoul 05029, Republic of Korea
| | - Yangmee Kim
- Department of Bioscience and Biotechnology, Konkuk University, Seoul 05029, Republic of Korea
| | - Wahn Soo Choi
- Department of Immunology, College of Medicine, Konkuk University, Chungju 27478, Republic of Korea
- Institute of Biomedical Sciences & Technology, Konkuk University, Seoul 05029, Republic of Korea
| |
Collapse
|
8
|
Van den Eynde R, Hertel F, Abakumov S, Krajnik B, Hugelier S, Auer A, Hellmeier J, Schlichthaerle T, Grattan RM, Lidke DS, Jungmann R, Leutenegger M, Vandenberg W, Dedecker P. Simultaneous multicolor fluorescence imaging using PSF splitting. Nat Methods 2024; 21:1801-1805. [PMID: 39242832 DOI: 10.1038/s41592-024-02383-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Accepted: 07/17/2024] [Indexed: 09/09/2024]
Abstract
We present a way to encode more information in fluorescence imaging by splitting the original point spread function (PSF), which offers broadband operation and compatibility with other PSF engineering modalities and existing analysis tools. We demonstrate the approach using the 'Circulator', an add-on that encodes the fluorophore emission band into the PSF, enabling simultaneous multicolor super-resolution and single-molecule microscopy using essentially the full field of view.
Collapse
Affiliation(s)
| | - Fabian Hertel
- Department of Chemistry, KU Leuven, Leuven, Belgium
- Department of Nuclear Medicine, University Hospital RWTH Aachen University, Aachen, Germany
| | | | - Bartosz Krajnik
- Department of Chemistry, KU Leuven, Leuven, Belgium
- Department of Experimental Physics, Wroclaw University of Science and Technology, Wroclaw, Poland
| | | | - Alexander Auer
- Max Planck Institute of Biochemistry, Martinsried, Germany
- Faculty of Physics and Center for Nanoscience, Ludwig Maximilian University, Munich, Germany
| | - Joschka Hellmeier
- Max Planck Institute of Biochemistry, Martinsried, Germany
- Faculty of Physics and Center for Nanoscience, Ludwig Maximilian University, Munich, Germany
| | - Thomas Schlichthaerle
- Max Planck Institute of Biochemistry, Martinsried, Germany
- Faculty of Physics and Center for Nanoscience, Ludwig Maximilian University, Munich, Germany
| | - Rachel M Grattan
- Department of Pathology and Comprehensive Cancer Center, University of New Mexico, Albuquerque, NM, USA
| | - Diane S Lidke
- Department of Pathology and Comprehensive Cancer Center, University of New Mexico, Albuquerque, NM, USA
| | - Ralf Jungmann
- Max Planck Institute of Biochemistry, Martinsried, Germany
- Faculty of Physics and Center for Nanoscience, Ludwig Maximilian University, Munich, Germany
| | | | | | | |
Collapse
|
9
|
Obeng B, Bennett LJ, West BE, Wagner DJ, Fleming PJ, Tasker MN, Lorenger MK, Smith DR, Systuk T, Plummer SM, Eom J, Paine MD, Frangos CT, Wilczek MP, Shim JK, Maginnis MS, Gosse JA. Antimicrobial cetylpyridinium chloride suppresses mast cell function by targeting tyrosine phosphorylation of Syk kinase. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.04.602096. [PMID: 39026716 PMCID: PMC11257455 DOI: 10.1101/2024.07.04.602096] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/20/2024]
Abstract
Cetylpyridinium chloride (CPC) is a quaternary ammonium antimicrobial used in numerous personal care products, human food, cosmetic products, and cleaning solutions. Yet, there is minimal published data on CPC effects on eukaryotes, immune signaling, and human health. Previously, we showed that low-micromolar CPC inhibits rat mast cell function by inhibiting antigen (Ag)-stimulated Ca 2+ mobilization, microtubule polymerization, and degranulation. In this study, we extend the findings to human mast cells (LAD2) and present data indicating that CPC's mechanism of action centers on its positively-charged quaternary nitrogen in its pyridinium headgroup. CPC's inhibitory effect is independent of signaling platform receptor architecture. Tyrosine phosphorylation events are a trigger of Ca 2+ mobilization necessary for degranulation. CPC inhibits global tyrosine phosphorylation in Ag-stimulated mast cells. Specifically, CPC inhibits tyrosine phosphorylation of specific key players Syk kinase and LAT, a substrate of Syk. In contrast, CPC does not affect Lyn kinase phosphorylation. Thus, CPC's root mechanism is electrostatic disruption of particular tyrosine phosphorylation events essential for signaling. This work outlines the biochemical mechanisms underlying the effects of CPC on immune signaling and allows the prediction of CPC effects on cell types, like T cells, that share similar signaling elements.
Collapse
|
10
|
Seo DY, Park JW, Kim SH, Oh SR, Han SB, Kwon OK, Ahn KS. Effect of Isoscopoletin on Cytokine Expression in HaCaT Keratinocytes and RBL-2H3 Basophils: Preliminary Study. Int J Mol Sci 2024; 25:6908. [PMID: 39000019 PMCID: PMC11240891 DOI: 10.3390/ijms25136908] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Revised: 06/19/2024] [Accepted: 06/20/2024] [Indexed: 07/14/2024] Open
Abstract
Isoscopoletin is a compound derived from various plants traditionally used for the treatment of skin diseases. However, there have been no reported therapeutic effects of isoscopoletin on atopic dermatitis (AD). AD is a chronic inflammatory skin disease, and commonly used treatments have side effects; thus, there is a need to identify potential natural candidate substances. In this study, we aimed to investigate whether isoscopoletin regulates the inflammatory mediators associated with AD in TNF-α/IFN-γ-treated HaCaT cells and PMA/ionomycin treated RBL-2H3 cells. We determined the influence of isoscopoletin on cell viability through an MTT assay and investigated the production of inflammatory mediators using ELISA and RT-qPCR. Moreover, we analyzed the transcription factors that regulate inflammatory mediators using Western blots and ICC. The results showed that isoscopoletin did not affect cell viability below 40 μM in either HaCaT or RBL-2H3 cells. Isoscopoletin suppressed the production of TARC/CCL17, MDC/CCL22, MCP-1/CCL2, IL-8/CXCL8, and IL-1β in TNF-α/IFN-γ-treated HaCaT cells and IL-4 in PMA/ionomycin-treated RBL-2H3 cells. Furthermore, in TNF-α/IFN-γ-treated HaCaT cells, the phosphorylation of signaling pathways, including MAPK, NF-κB, STAT, and AKT/PKB, increased but was decreased by isoscopoletin. In PMA/ionomycin-treated RBL-2H3 cells, the activation of signaling pathways including PKC, MAPK, and AP-1 increased but was decreased by isoscopoletin. In summary, isoscopoletin reduced the production of inflammatory mediators by regulating upstream transcription factors in TNF-α/IFN-γ-treated HaCaT cells and PMA/ionomycin-treated RBL-2H3 cells. Therefore, we suggest that isoscopoletin has the potential for a therapeutic effect, particularly in skin inflammatory diseases such as AD, by targeting keratinocytes and basophils.
Collapse
Affiliation(s)
- Da-Yun Seo
- College of Pharmacy, Chungbuk National University, Cheongju 28160, Republic of Korea; (D.-Y.S.); (S.-B.H.)
- Natural Medicine Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju 28116, Republic of Korea; (S.-H.K.); (S.-R.O.)
| | - Ji-Won Park
- Practical Research Division, Honam National Institute of Biological Resources (HNIBR), Mokpo 58762, Republic of Korea;
| | - Seung-Ho Kim
- Natural Medicine Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju 28116, Republic of Korea; (S.-H.K.); (S.-R.O.)
| | - Sei-Ryang Oh
- Natural Medicine Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju 28116, Republic of Korea; (S.-H.K.); (S.-R.O.)
| | - Sang-Bae Han
- College of Pharmacy, Chungbuk National University, Cheongju 28160, Republic of Korea; (D.-Y.S.); (S.-B.H.)
| | - Ok-Kyoung Kwon
- Natural Medicine Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju 28116, Republic of Korea; (S.-H.K.); (S.-R.O.)
| | - Kyung-Seop Ahn
- Natural Medicine Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju 28116, Republic of Korea; (S.-H.K.); (S.-R.O.)
| |
Collapse
|
11
|
Capener JL, Vasta JD, Katis VL, Michaud A, Beck MT, Daglish SCD, Cohen-Kedar S, Barda ES, Howell S, Dotan I, Robers MB, Axtman AD, Bashore FM. Development of SYK NanoBRET Cellular Target Engagement Assays for Gain-of-Function Variants. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.12.598544. [PMID: 38915605 PMCID: PMC11195201 DOI: 10.1101/2024.06.12.598544] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/26/2024]
Abstract
Spleen tyrosine kinase (SYK) is a non-receptor tyrosine kinase that is activated by phosphorylation events downstream of FcR, B-cell and T-cell receptors, integrins, and C-type lectin receptors. When the tandem Src homology 2 (SH2) domains of SYK bind to phosphorylated immunoreceptor tyrosine-based activation motifs (pITAMs) contained within these immunoreceptors, or when SYK is phosphorylated in interdomain regions A and B, SYK is activated. SYK gain-of-function (GoF) variants were previously identified in six patients that had higher levels of phosphorylated SYK and phosphorylated downstream proteins JNK and ERK. Furthermore, the increased SYK activation resulted in the clinical manifestation of immune dysregulation, organ inflammation, and a predisposition for lymphoma. The knowledge that the SYK GoF variants have enhanced activity was leveraged to develop a SYK NanoBRET cellular target engagement assay in intact live cells with constructs for the SYK GoF variants. Herein, we developed a potent SYK-targeted NanoBRET tracer using a SYK donated chemical probe, MRL-SYKi, that enabled a NanoBRET cellular target engagement assay for SYK GoF variants, SYK(S550Y), SYK(S550F), and SYK(P342T). We determined that ATP-competitive SYK inhibitors bind potently to these SYK variants in intact live cells. Additionally, we demonstrated that MRL-SYKi can effectively reduce the catalytic activity of SYK variants, and the phosphorylation levels of SYK(S550Y) in an epithelial cell line (SW480) stably expressing SYK(S550Y).
Collapse
Affiliation(s)
- Jacob L. Capener
- Structural Genomics Consortium, Division of Chemical Biology and Medicinal Chemistry, Eshelman School of Pharmacy, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - James D. Vasta
- Promega Corporation, 2800 Woods Hollow Road, Madison, WI 53711, USA
| | - Vittorio L. Katis
- ARUK Oxford Drug Discovery Institute, Centre for Medicines Discovery, Nuffield Department of Medicine, University of Oxford, Oxford OX3 7FZ, UK
| | - Ani Michaud
- Promega Corporation, 2800 Woods Hollow Road, Madison, WI 53711, USA
| | - Michael T. Beck
- Promega Corporation, 2800 Woods Hollow Road, Madison, WI 53711, USA
| | - Sabrina C. D. Daglish
- Structural Genomics Consortium, Division of Chemical Biology and Medicinal Chemistry, Eshelman School of Pharmacy, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Sarit Cohen-Kedar
- Division of Gastroenterology, Rabin Medical Center, Petah-Tikva, Israel
- Felsenstein Medical Research Center, Faculty of Medical & Health Sciences, Tel-Aviv University, Tel-Aviv, Israel
| | - Efrat Shaham Barda
- Division of Gastroenterology, Rabin Medical Center, Petah-Tikva, Israel
- Felsenstein Medical Research Center, Faculty of Medical & Health Sciences, Tel-Aviv University, Tel-Aviv, Israel
| | - Stefanie Howell
- Structural Genomics Consortium, Division of Chemical Biology and Medicinal Chemistry, Eshelman School of Pharmacy, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Iris Dotan
- Division of Gastroenterology, Rabin Medical Center, Petah-Tikva, Israel
- Felsenstein Medical Research Center, Faculty of Medical & Health Sciences, Tel-Aviv University, Tel-Aviv, Israel
| | | | - Alison D. Axtman
- Structural Genomics Consortium, Division of Chemical Biology and Medicinal Chemistry, Eshelman School of Pharmacy, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Frances M. Bashore
- Structural Genomics Consortium, Division of Chemical Biology and Medicinal Chemistry, Eshelman School of Pharmacy, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| |
Collapse
|
12
|
Dibaji H, Kazemi Nasaban Shotorban A, Grattan RM, Lucero S, Schodt DJ, Lidke KA, Petruccelli J, Lidke DS, Liu S, Chakraborty T. Axial de-scanning using remote focusing in the detection arm of light-sheet microscopy. Nat Commun 2024; 15:5019. [PMID: 38866746 PMCID: PMC11169345 DOI: 10.1038/s41467-024-49291-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Accepted: 05/31/2024] [Indexed: 06/14/2024] Open
Abstract
Rapid, high-resolution volumetric imaging without moving heavy objectives or disturbing delicate samples remains challenging. Pupil-matched remote focusing offers a promising solution for high NA systems, but the fluorescence signal's incoherent and unpolarized nature complicates its application. Thus, remote focusing is mainly used in the illumination arm with polarized laser light to improve optical coupling. Here, we introduce a novel optical design that can de-scan the axial focus movement in the detection arm of a microscope. Our method splits the fluorescence signal into S and P-polarized light, lets them pass through the remote focusing module separately, and combines them with the camera. This allows us to use only one focusing element to perform aberration-free, multi-color, volumetric imaging without (a) compromising the fluorescent signal and (b) needing to perform sample/detection-objective translation. We demonstrate the capabilities of this scheme by acquiring fast dual-color 4D (3D space + time) image stacks with an axial range of 70 μm and camera-limited acquisition speed. Owing to its general nature, we believe this technique will find its application in many other microscopy techniques that currently use an adjustable Z-stage to carry out volumetric imaging, such as confocal, 2-photon, and light sheet variants.
Collapse
Affiliation(s)
- Hassan Dibaji
- Department of Physics and Astronomy, University of New Mexico, Albuquerque, NM, USA
| | | | - Rachel M Grattan
- Comprehensive Cancer Center, University of New Mexico Health Sciences Center, Albuquerque, NM, USA
- Department of Pathology, University of New Mexico Health Science Center, Albuquerque, NM, USA
| | - Shayna Lucero
- Comprehensive Cancer Center, University of New Mexico Health Sciences Center, Albuquerque, NM, USA
- Department of Pathology, University of New Mexico Health Science Center, Albuquerque, NM, USA
| | - David J Schodt
- Department of Physics and Astronomy, University of New Mexico, Albuquerque, NM, USA
| | - Keith A Lidke
- Department of Physics and Astronomy, University of New Mexico, Albuquerque, NM, USA
- Comprehensive Cancer Center, University of New Mexico Health Sciences Center, Albuquerque, NM, USA
| | - Jonathan Petruccelli
- Department of Physics, University at Albany-State University of NewYork, Albany, NY, USA
| | - Diane S Lidke
- Comprehensive Cancer Center, University of New Mexico Health Sciences Center, Albuquerque, NM, USA
- Department of Pathology, University of New Mexico Health Science Center, Albuquerque, NM, USA
| | - Sheng Liu
- Department of Physics and Astronomy, University of New Mexico, Albuquerque, NM, USA
| | - Tonmoy Chakraborty
- Department of Physics and Astronomy, University of New Mexico, Albuquerque, NM, USA.
- Comprehensive Cancer Center, University of New Mexico Health Sciences Center, Albuquerque, NM, USA.
| |
Collapse
|
13
|
Furiness KN, El Ansari YS, Oettgen HC, Kanagaratham C. Allergen-specific IgA and IgG antibodies as inhibitors of mast cell function in food allergy. FRONTIERS IN ALLERGY 2024; 5:1389669. [PMID: 38919913 PMCID: PMC11196826 DOI: 10.3389/falgy.2024.1389669] [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: 02/21/2024] [Accepted: 05/21/2024] [Indexed: 06/27/2024] Open
Abstract
Food allergy, a group of adverse immune responses to normally innocuous food protein antigens, is an increasingly prevalent public health issue. The most common form is IgE-mediated food allergy in which food antigen-induced crosslinking of the high-affinity IgE-receptor, FcεRI, on the surface of mast cells triggers the release of inflammatory mediators that contribute to a wide range of clinical manifestations, including systemic anaphylaxis. Mast cells also play a critical function in adaptive immunity to foods, acting as adjuvants for food-antigen driven Th2 cell responses. While the diagnosis and treatment of food allergy has improved in recent years, no curative treatments are currently available. However, there is emerging evidence to suggest that both allergen-specific IgA and IgG antibodies can counter the activating effects of IgE antibodies on mast cells. Most notably, both antigen-specific IgA and IgG antibodies are induced in the course of oral immunotherapy. In this review, we highlight the role of mast cells in food allergy, both as inducers of immediate hypersensitivity reactions and as adjuvants for type 2 adaptive immune responses. Furthermore, we summarize current understanding of the immunomodulatory effects of antigen-specific IgA and IgG antibodies on IgE-induced mast cell activation and effector function. A more comprehensive understanding of the regulatory role of IgA and IgG in food allergy may provide insights into physiologic regulation of immune responses to ingested antigens and could seed novel strategies to treat allergic disease.
Collapse
Affiliation(s)
- Kameryn N. Furiness
- Division of Immunology, Department of Pediatrics, Boston Children’s Hospital, Boston, MA, United States
| | - Yasmeen S. El Ansari
- Division of Immunology, Department of Pediatrics, Boston Children’s Hospital, Boston, MA, United States
- Institute of Laboratory Medicine, Philipps University Marburg, Marburg, Germany
| | - Hans C. Oettgen
- Division of Immunology, Department of Pediatrics, Boston Children’s Hospital, Boston, MA, United States
- Department of Pediatrics, Harvard Medical School, Boston, MA, United States
| | - Cynthia Kanagaratham
- Division of Immunology, Department of Pediatrics, Boston Children’s Hospital, Boston, MA, United States
- Department of Pediatrics, Harvard Medical School, Boston, MA, United States
| |
Collapse
|
14
|
Shaik GM, Khan MS. Betulinic Acid Potentiates Mast Cell Degranulation by Compromising Cell Membrane Integrity and Without Involving Fcεri Receptors. Immunol Invest 2024; 53:695-711. [PMID: 38504489 DOI: 10.1080/08820139.2024.2329990] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/21/2024]
Abstract
Mast cells play important role in acquired and natural immunity making these favorable therapeutic targets in various inflammatory diseases. Here we observed that, pentacyclic tri terpenoid betulinic acid (BA) treatment resulted in a significantly high number (9%) of cells positive for Hoechst and negative for annexin-V indicating that BA could interfere with plasma membrane integrity. The degranulation of both activated and non-activated mast cells was enhanced upon treatment with BA. The pre-treatment of BA had remarkable effect on calcium response in activated mast cells which showed increased calcium influx relative compared to untreated cells. The results also showed potentially less migration of BA treated mast cells signifying the possible effect of BA on cell membrane. BA treatment resulted in a significant increase in mRNA levels of IL-13 while as mRNA levels of other target cytokines, IL-6 and TNF-α seem to be not affected. Moreover, there was global Increase in phosphorylation of signaling proteins and no significant change in phosphorylation of FcεRI receptors indicating that the effect of BA was independent of signaling cascade or FcεRI receptor mediated mast cell aggregation. Overall, these results portray BA potentiates mast cell effector functions by compromising the membrane integrity and independent of FcεRI involvement.
Collapse
Affiliation(s)
- Gouse M Shaik
- Department of Biochemistry, College of Science, King Saud University, Riyadh, Saudi Arabia
| | - Mohd Shahnawaz Khan
- Department of Biochemistry, College of Science, King Saud University, Riyadh, Saudi Arabia
| |
Collapse
|
15
|
Maheshika Kumari Jayasinghe A, Yang HW, Gedara Isuru Sandanuwan Kirindage K, Jung K, Je JG, Wang L, Kim KN, Ahn G. Fucosterol isolated from Sargassum horneri attenuates allergic responses in immunoglobulin E/bovine serum albumin-stimulated mast cells and passive cutaneous anaphylaxis in mice. Int Immunopharmacol 2024; 131:111851. [PMID: 38492337 DOI: 10.1016/j.intimp.2024.111851] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Revised: 02/29/2024] [Accepted: 03/10/2024] [Indexed: 03/18/2024]
Abstract
Allergic diseases have become a serious problem worldwide and occur when the immune system overreacts to stimuli. Sargassum horneri is an edible marine brown alga with pharmacological relevance in treating various allergy-related conditions. Therefore, this study aimed to investigate the effect of fucosterol (FST) isolated from S. horneri on immunoglobulin E(IgE)/bovine serum albumin (BSA)-stimulated allergic reactions in mouse bone marrow-derived cultured mast cells (BMCMCs) and passive cutaneous anaphylaxis (PCA) in BALB/c mice. The in silico analysis results revealed the binding site modulatory potential of FST on the IgE and IgE-FcεRI complex. The findings of the study revealed that FST significantly suppressed the degranulation of IgE/BSA-stimulated BMCMCs by inhibiting the release of β-hexosaminidase and histamine in a dose-dependent manner. In addition, FST effectively decreased the expression of FcεRI on the surface of BMCMCs and its IgE binding. FST dose-dependently downregulated the expression of allergy-related cytokines (interleukin (IL)-4, -5, -6, -13, tumor necrosis factor (TNF)-α, and a chemokine (thymus and activation-regulated chemokine (TARC)) by suppressing the activation of nuclear factor-κB (NF-κB) and Syk-LAT-ERK-Gab2 signaling in IgE/BSA-stimulated BMCMCs. As per the histological analysis results of the in vivo studies with IgE-mediated PCA in BALB/c mice, FST treatment effectively attenuated the PCA reactions. These findings suggest that FST has an immunopharmacological potential as a naturally available bioactive compound for treating allergic reactions.
Collapse
Affiliation(s)
| | - Hye-Won Yang
- Department of Marine Life Science, Jeju National University, Jeju 63243, Republic of Korea.
| | | | - Kyungsook Jung
- Functional Biomaterials Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Jeongeup-si 56212, Republic of Korea.
| | - Jun-Geon Je
- Department of Marine Life Science, Jeju National University, Jeju 63243, Republic of Korea.
| | - Lei Wang
- College of Food Science and Engineering, Ocean University of China, Qingdao 266003, China.
| | - Kil-Nam Kim
- Chuncheon Center, Korea Basic Science Institute (KBSI), Chuncheon 24341, Republic of Korea.
| | - Ginnae Ahn
- Department of Food Technology and Nutrition, Chonnam National University, Yeosu 59626, Republic of Korea; Department of Marine Bio-Food Sciences, Chonnam National University, Yeosu 59626, Republic of Korea.
| |
Collapse
|
16
|
Dibaji H, Nasaban Shotorban AK, Grattan RM, Lucero S, Schodt DJ, Lidke KA, Petruccelli J, Lidke DS, Liu S, Chakraborty T. Axial de-scanning using remote focusing in the detection arm of light-sheet microscopy. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.09.07.556729. [PMID: 38659774 PMCID: PMC11042218 DOI: 10.1101/2023.09.07.556729] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/26/2024]
Abstract
The ability to image at high speeds is necessary for biological imaging to capture fast-moving or transient events or to efficiently image large samples. However, due to the lack of rigidity of biological specimens, carrying out fast, high-resolution volumetric imaging without moving and agitating the sample has been a challenging problem. Pupil-matched remote focusing has been promising for high NA imaging systems with their low aberrations and wavelength independence, making it suitable for multicolor imaging. However, owing to the incoherent and unpolarized nature of the fluorescence signal, manipulating this emission light through remote focusing is challenging. Therefore, remote focusing has been primarily limited to the illumination arm, using polarized laser light to facilitate coupling in and out of the remote focusing optics. Here, we introduce a novel optical design that can de-scan the axial focus movement in the detection arm of a microscope. Our method splits the fluorescence signal into S and P-polarized light, lets them pass through the remote focusing module separately, and combines them with the camera. This allows us to use only one focusing element to perform aberration-free, multi-color, volumetric imaging without (a) compromising the fluorescent signal and (b) needing to perform sample/detection-objective translation. We demonstrate the capabilities of this scheme by acquiring fast dual-color 4D (3D space + time) image stacks with an axial range of 70 μm and camera-limited acquisition speed. Owing to its general nature, we believe this technique will find its application in many other microscopy techniques that currently use an adjustable Z-stage to carry out volumetric imaging, such as confocal, 2-photon, and light sheet variants.
Collapse
|
17
|
Jayasinghe AMK, Kirindage KGIS, Kim SH, Lee S, Kim KN, Kim EA, Heo SJ, Ahn G. Leaves and pseudostems extract of Curcuma longa attenuates immunoglobulin E/bovine serum albumin-stimulated bone marrow-derived cultured mast cell activation and passive cutaneous anaphylaxis in BALB/c mice. JOURNAL OF ETHNOPHARMACOLOGY 2024; 321:117529. [PMID: 38042384 DOI: 10.1016/j.jep.2023.117529] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Revised: 11/24/2023] [Accepted: 11/27/2023] [Indexed: 12/04/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Curcuma longa, known as turmeric, is an herbaceous perennial plant belonging to the genus Curcuma. It is dispersed throughout tropical and subtropical regions worldwide. Since ancient times, turmeric has been used as an ethnomedicinal plant in the Ayurvedic system, particularly in Asian countries. Rhizomes of turmeric possess several pharmacological properties that give high value as a medicinal remedy for treating a range of conditions, including inflammation, pain, allergies, and digestive issues. Moreover, turmeric leaves and pseudostems also contain a variety of health-enhancing secondary metabolites, such as curcumin, flavonoids, and other phenolic compounds, which exhibit anti-inflammatory, antitumor, antibacterial, and antioxidant properties. AIM OF THE STUDY Allergic diseases are a group of immune-mediated disorders mainly caused by an immunoglobulin E (IgE)-dependent immunological response to an innocuous allergen. Therefore, this study aimed to investigate the effect of leaves and pseudostems extract of turmeric (TLSWE-8510) on IgE/bovine serum albumin (BSA)-stimulated allergic responses in mouse bone marrow-derived cultured mast cells (BMCMCs) and passive cutaneous anaphylaxis (PCA) in BALB/c mice. MATERIALS AND METHODS The effect of TLSWE-8510 on mast cell degranulation has been evaluated by investigating the release of β-hexosaminidase and histamine in IgE/BSA-stimulated BMCMCs. Additionally, anti-allergic properties of TLSWE-8510 on IgE/BSA-stimulated BMCMCs were investigated using suppression of nuclear factor-kappa B (NF-κB), and spleen tyrosine kinase (Syk)-linker for T-cell activation (LAT)-extracellular-signal-regulated kinase (ERK)-GRB2 associated binding protein 2 (Gab2) signaling pathway and downregulation of allergy-related cytokines and chemokines expression. Furthermore, in vivo, studies were conducted using IgE-mediated PCA in BALB/c mice. RESULTS TLSWE-8510 treatment significantly inhibited the degranulation of IgE/BSA-stimulated BMCMCs by inhibiting the release of β-hexosaminidase and histamine dose-dependently. Additionally, TLSWE-8510 reduced the expression of high-affinity IgE receptors (Fc epsilon receptor I-FcεRI) on the surface of BMCMCs and the binding of IgE to FcεRI. Besides, the expression of cytokines and chemokines is triggered by IgE/BSA stimulation via activating the allergy-related signaling pathways. TLSWE-8510 dose-dependently downregulated the mRNA expression and the production of allergy-related cytokines (interleukin (IL)-1β, IL-3, IL-4, IL-5, IL-6, IL-13, tumor necrosis factor (TNF)-α, and interferon (IFN)-γ), and chemokines (thymus and activation-regulated chemokine (TARC), and regulated upon activation, normal T cell expressed and secreted (RANTES)) by regulating the phosphorylation of downstream signaling molecules, NF-κB, and Syk, LAT, ERK and Gab2 in IgE/BSA-stimulated BMCMCs. Moreover, PCA reaction in IgE/BSA-stimulated BALB/c mice ears was effectively decreased by TLSWE-8510 treatment in a dose-dependent manner. CONCLUSIONS These results collectively demonstrated that TLSWE-8510 suppressed mast cell degranulation by inhibiting the release of chemical mediators related to allergies. TLSWE-8510 downregulated the allergy-related cytokines and chemokines expression and phosphorylation of downstream signaling molecules in IgE/BSA-stimulated BMCMCs. Furthermore, in vivo studies with IgE-mediated PCA reaction in the BALB/c mice ears were attenuated by TLSWE-8510 treatment. These findings revealed that TLSWE-8510 has the potential as a therapeutic agent for the treatment of allergic diseases.
Collapse
Affiliation(s)
| | | | - Sun-Hyung Kim
- French Korea Aromatics Co., Ltd., Yongin-si, Gyeonggi-do, Republic of Korea.
| | - Seok Lee
- French Korea Aromatics Co., Ltd., Yongin-si, Gyeonggi-do, Republic of Korea.
| | - Kil-Nam Kim
- Gwangju Center, Korea Basic Science Institute (KBSI), Gwangju, 61751, Republic of Korea.
| | - Eun-A Kim
- Jeju International Marine Science Center for Research & Education, Korea Institute of Ocean Science & Technology (KIOST), Jeju, 63349, Republic of Korea.
| | - Soo-Jin Heo
- Jeju International Marine Science Center for Research & Education, Korea Institute of Ocean Science & Technology (KIOST), Jeju, 63349, Republic of Korea.
| | - Ginnae Ahn
- Department of Food Technology and Nutrition, Chonnam National University, Yeosu, 59626, Republic of Korea; Department of Marine Bio-Food Sciences, Chonnam National University, Yeosu, 59626, Republic of Korea.
| |
Collapse
|
18
|
Choi KS, Shin TS, Ahn G, Kim SH, Chun J, Lee M, Kim DH, Choi HG, Lee KD, Shim SY. Bioactivity-Guided Fraction from Viscera of Abalone, Haliotis discus hannai Suppresses Cellular Basophils Activation and Anaphylaxis in Mice. J Microbiol Biotechnol 2024; 34:379-386. [PMID: 38037338 PMCID: PMC10940748 DOI: 10.4014/jmb.2310.10015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Revised: 10/31/2023] [Accepted: 11/08/2023] [Indexed: 12/02/2023]
Abstract
Basophils and mast cells are specialized effector cells in allergic reactions. Haliotis discus hannai (abalone), is valuable seafood. Abalone male viscera, which has a brownish color and has not been previously reported to show anti-allergic activities, was extracted with acetone. Six different acetone/hexane fractions (0, 10, 20, 30, 40, and 100%) were obtained using a silica column via β-hexosaminidase release inhibitory activity-guided selection in phorbol myristate acetate and a calcium ionophore, A23187 (PMACI)-induced human basophils, KU812F cells. The 40% acetone/hexane fraction (A40) exhibited the strongest inhibition of PMACI-induced-β-hexosaminidase release. This fraction dose-dependently inhibited reactive oxygen species (ROS) production and calcium mobilization without cytotoxicity. Western blot analysis revealed that A40 down-regulated PMACI-induced MAPK (ERK 1/2, p-38, and JNK) phosphorylation, and the NF-κB translocation from the cytosol to membrane. Moreover, A40 inhibited PMACI-induced interleukin (IL)-1β, IL-6, and IL-8 production. Anti-allergic activities of A40 were confirmed based on inhibitory effects on IL-4 and tumor necrosis factor alpha (TNF-α) production in compound (com) 48/80-induced rat basophilic leukemia (RBL)-2H3 cells. A40 inhibited β-hexosaminidase release and cytokine production such as IL-4 and TNF-α produced by com 48/80-stimulated RBL-2H3 cells. Furthermore, it's fraction attenuated the IgE/DNP-induced passive cutaneous anaphylaxis (PCA) reaction in the ears of BALB/c mice. Our results suggest that abalone contains the active fraction, A40 is a potent therapeutic and functional material to treat allergic diseases.
Collapse
Affiliation(s)
- Kap Seong Choi
- Department of Food Science and Technology, Sunchon National University, Suncheon 57922, Republic of Korea
| | - Tai-Sun Shin
- Division of Food and Nutrition, Chonnam National University, Gwangju 61186, Republic of Korea
| | - Ginnae Ahn
- Department of Marine Bio-Food Sciences, Chonnam National University, Yeosu 59626, Republic of Korea
| | - Shin Hye Kim
- Division of Food and Nutrition, Chonnam National University, Gwangju 61186, Republic of Korea
| | - Jiyeon Chun
- Department of Food Science and Technology, Sunchon National University, Suncheon 57922, Republic of Korea
| | - Mina Lee
- College of Pharmacy and Research Institute of Life and Pharmaceutical Sciences, Sunchon National University, Suncheon 57922, Republic of Korea
| | - Dae Heon Kim
- Department of Biomedical Science, Sunchon National University, Suncheon 57922, Republic of Korea
| | - Han-Gil Choi
- Faculty of Biological Science and Institute for Environmental Science, Wonkwang University, Iksan 54538, Republic of Korea
| | - Kyung-Dong Lee
- Department of Companion animal industry, College of Health & Welfare, Dongshin University. Naju 58245, Republic of Korea
| | - Sun-Yup Shim
- Department of Food Science and Technology, Sunchon National University, Suncheon 57922, Republic of Korea
| |
Collapse
|
19
|
Bashore FM, Katis VL, Du Y, Sikdar A, Wang D, Bradshaw WJ, Rygiel KA, Leisner TM, Chalk R, Mishra S, Williams CA, Gileadi O, Brennan PE, Wiley JC, Gockley J, Cary GA, Carter GW, Young JE, Pearce KH, Fu H, the Emory-Sage-SGC TREAT-AD Center, Axtman AD. Characterization of covalent inhibitors that disrupt the interaction between the tandem SH2 domains of SYK and FCER1G phospho-ITAM. PLoS One 2024; 19:e0293548. [PMID: 38359047 PMCID: PMC10868801 DOI: 10.1371/journal.pone.0293548] [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: 08/02/2023] [Accepted: 10/15/2023] [Indexed: 02/17/2024] Open
Abstract
RNA sequencing and genetic data support spleen tyrosine kinase (SYK) and high affinity immunoglobulin epsilon receptor subunit gamma (FCER1G) as putative targets to be modulated for Alzheimer's disease (AD) therapy. FCER1G is a component of Fc receptor complexes that contain an immunoreceptor tyrosine-based activation motif (ITAM). SYK interacts with the Fc receptor by binding to doubly phosphorylated ITAM (p-ITAM) via its two tandem SH2 domains (SYK-tSH2). Interaction of the FCER1G p-ITAM with SYK-tSH2 enables SYK activation via phosphorylation. Since SYK activation is reported to exacerbate AD pathology, we hypothesized that disruption of this interaction would be beneficial for AD patients. Herein, we developed biochemical and biophysical assays to enable the discovery of small molecules that perturb the interaction between the FCER1G p-ITAM and SYK-tSH2. We identified two distinct chemotypes using a high-throughput screen (HTS) and orthogonally assessed their binding. Both chemotypes covalently modify SYK-tSH2 and inhibit its interaction with FCER1G p-ITAM, however, these compounds lack selectivity and this limits their utility as chemical tools.
Collapse
Affiliation(s)
- Frances M. Bashore
- Structural Genomics Consortium, Division of Chemical Biology and Medicinal Chemistry, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States of America
| | - Vittorio L. Katis
- Nuffield Department of Medicine, Centre for Medicines Discovery, ARUK Oxford Drug Discovery Institute, University of Oxford, Headington, Oxford, United Kingdom
| | - Yuhong Du
- Department of Pharmacology and Chemical Biology, School of Medicine, Emory University, Atlanta, GA, United States of America
- Emory Chemical Biology Discovery Center, School of Medicine, Emory University, Atlanta, GA, United States of America
| | - Arunima Sikdar
- Division of Chemical Biology and Medicinal Chemistry, Center for Integrative Chemical Biology and Drug Discovery, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States of America
| | - Dongxue Wang
- Department of Pharmacology and Chemical Biology, School of Medicine, Emory University, Atlanta, GA, United States of America
- Emory Chemical Biology Discovery Center, School of Medicine, Emory University, Atlanta, GA, United States of America
| | - William J. Bradshaw
- Nuffield Department of Medicine, Centre for Medicines Discovery, ARUK Oxford Drug Discovery Institute, University of Oxford, Headington, Oxford, United Kingdom
| | - Karolina A. Rygiel
- Nuffield Department of Medicine, Centre for Medicines Discovery, ARUK Oxford Drug Discovery Institute, University of Oxford, Headington, Oxford, United Kingdom
| | - Tina M. Leisner
- Division of Chemical Biology and Medicinal Chemistry, Center for Integrative Chemical Biology and Drug Discovery, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States of America
| | - Rod Chalk
- Nuffield Department of Medicine, Centre for Medicines Discovery, ARUK Oxford Drug Discovery Institute, University of Oxford, Headington, Oxford, United Kingdom
| | - Swati Mishra
- Department of Laboratory Medicine and Pathology, School of Medicine, University of Washington, Seattle, WA, United States of America
- Institute for Stem Cell and Regenerative Medicine, School of Medicine, University of Washington, Seattle, WA, United States of America
| | - C. Andrew Williams
- Department of Laboratory Medicine and Pathology, School of Medicine, University of Washington, Seattle, WA, United States of America
- Institute for Stem Cell and Regenerative Medicine, School of Medicine, University of Washington, Seattle, WA, United States of America
| | - Opher Gileadi
- Nuffield Department of Medicine, Centre for Medicines Discovery, ARUK Oxford Drug Discovery Institute, University of Oxford, Headington, Oxford, United Kingdom
| | - Paul E. Brennan
- Nuffield Department of Medicine, Centre for Medicines Discovery, ARUK Oxford Drug Discovery Institute, University of Oxford, Headington, Oxford, United Kingdom
| | | | - Jake Gockley
- Sage Bionetworks, Seattle, WA, United States of America
| | - Gregory A. Cary
- The Jackson Laboratory for Mammalian Genetics, Bar Harbor, ME, United States of America
| | - Gregory W. Carter
- The Jackson Laboratory for Mammalian Genetics, Bar Harbor, ME, United States of America
| | - Jessica E. Young
- Department of Laboratory Medicine and Pathology, School of Medicine, University of Washington, Seattle, WA, United States of America
- Institute for Stem Cell and Regenerative Medicine, School of Medicine, University of Washington, Seattle, WA, United States of America
| | - Kenneth H. Pearce
- Division of Chemical Biology and Medicinal Chemistry, Center for Integrative Chemical Biology and Drug Discovery, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States of America
| | - Haian Fu
- Department of Pharmacology and Chemical Biology, School of Medicine, Emory University, Atlanta, GA, United States of America
- Emory Chemical Biology Discovery Center, School of Medicine, Emory University, Atlanta, GA, United States of America
| | | | - Alison D. Axtman
- Structural Genomics Consortium, Division of Chemical Biology and Medicinal Chemistry, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States of America
| |
Collapse
|
20
|
Mansouri M, ElHaddoumi G, Kandoussi I, Belyamani L, Ibrahimi A, El Hafidi N. Syk protein inhibitors treatment for the allergic symptoms associated with hyper immunoglobulin E syndromes: A focused on a computational approach. Int J Immunopathol Pharmacol 2024; 38:3946320241282030. [PMID: 39241232 PMCID: PMC11380138 DOI: 10.1177/03946320241282030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/08/2024] Open
Abstract
Background: Mutations in the Spleen tyrosine kinase (Syk) protein have significant implications for its function and response to treatments. Understanding these mutations and identifying new inhibitors can lead to more effective therapies for conditions like autosomal dominant hyper-IgE syndrome (AD-HIES) and related immunological disorders. Objective: To investigate the impact of mutations in the Syk protein on its function and response to reference treatments, and to explore new inhibitors tailored to the mutational profile of Syk. Methods: We collected and analyzed mutations affecting the Syk protein to assess their functional impact. We screened 94 deleterious mutations in the kinase domain using molecular docking techniques. A library of 997 compounds with potential inhibitory activity against Syk was filtered based on Lipinski and Veber rules and toxicity assessments. We evaluated the binding affinity of reference inhibitors and 14 eligible compounds against wild-type and mutant Syk proteins. Molecular dynamics simulations were conducted to evaluate the interaction of Syk protein complexes with the reference inhibitor and potential candidate inhibitors. Results: Among the analyzed mutations, 60.5% were identified as deleterious, underscoring their potential impact on cellular processes. Virtual screening identified three potential inhibitors (IDs: 118558008, 118558000, and 118558092) with greater therapeutic potential than reference treatments, meeting all criteria and exhibiting lower IC50 values. Ligand 1 (ID: 118558000) demonstrated the most stable binding, favorable compactness, and extensive interaction with solvents. A 3D pharmacophore model was constructed, identifying structural features common to these inhibitors. Conclusion: This study found that 60.5% of reported mutations affecting the Syk protein are deleterious. Virtual screening revealed three top potential inhibitors, with ligand 1 (ID: 118558000) showing the most stable binding and favorable interactions. These inhibitors hold promise for more effective therapies targeting Syk-mediated signaling pathways. The pharmacophore model provides valuable insights for developing targeted therapies for AD-HIES and related disorders, offering hope for patients suffering from Hyper IgE syndrome with allergic symptoms.
Collapse
Affiliation(s)
- Mariam Mansouri
- Biotechnology Lab (MedBiotech), Bioinova Research Center, Medical and Pharmacy School, Mohammed V University, Rabat, Morocco
| | - Ghyzlane ElHaddoumi
- Biotechnology Lab (MedBiotech), Bioinova Research Center, Medical and Pharmacy School, Mohammed V University, Rabat, Morocco
| | - Ilham Kandoussi
- Biotechnology Lab (MedBiotech), Bioinova Research Center, Medical and Pharmacy School, Mohammed V University, Rabat, Morocco
| | - Lahcen Belyamani
- Mohammed VI Center of Research and Innovation (CM6), Rabat, Morocco
- Mohammed VI University of Health Sciences (UM6SS), Casablanca, Morocco
- Emergency Department, Military Hospital Mohammed V, Medical and Pharmacy School, Mohammed V University, Rabat, Morocco
| | - Azeddine Ibrahimi
- Biotechnology Lab (MedBiotech), Bioinova Research Center, Medical and Pharmacy School, Mohammed V University, Rabat, Morocco
| | - Naima El Hafidi
- Biotechnology Lab (MedBiotech), Bioinova Research Center, Medical and Pharmacy School, Mohammed V University, Rabat, Morocco
- Mohammed VI Center of Research and Innovation (CM6), Rabat, Morocco
- Division of Pediatric Immunoallergology and Infectious Diseases, Children University Hospital, Ibn Sina University, Rabat, Morocco
| |
Collapse
|
21
|
Wang Z, Qu S, Yuan J, Tian W, Xu J, Tao R, Sun S, Lu T, Tang W, Zhu Y. Review and prospects of targeted therapies for Spleen tyrosine kinase (SYK). Bioorg Med Chem 2023; 96:117514. [PMID: 37984216 DOI: 10.1016/j.bmc.2023.117514] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2023] [Revised: 10/30/2023] [Accepted: 10/31/2023] [Indexed: 11/22/2023]
Abstract
Spleen tyrosine kinase (SYK) is a non-receptor tyrosine kinase. The dysregulation of SYK is closely related to the occurrence and development of allergic diseases, autoimmune diseases and cancer. SYK has become an attractive target for drug discovery due to its important biological functions. This article reviews the biological function of SYK, the relationship between SYK and disease, and therapies targeting SYK. In addition, inspired by new technologies such as proteolysis targeting chimeras (PROTACs) and phosphatase recruiting chimeras (PHORCs), we propose the development of new therapeutic approaches for targeting SYK, such as SYK PROTACs and SYK PHORCs, which may overcome deficiencies of existing methods.
Collapse
Affiliation(s)
- Zhaozhao Wang
- School of Science, China Pharmaceutical University, 639 Longmian Avenue, Nanjing 211198, PR China
| | - Shu Qu
- School of Science, China Pharmaceutical University, 639 Longmian Avenue, Nanjing 211198, PR China
| | - Jiahao Yuan
- School of Science, China Pharmaceutical University, 639 Longmian Avenue, Nanjing 211198, PR China
| | - Wen Tian
- School of Science, China Pharmaceutical University, 639 Longmian Avenue, Nanjing 211198, PR China
| | - Jinglei Xu
- School of Science, China Pharmaceutical University, 639 Longmian Avenue, Nanjing 211198, PR China
| | - Rui Tao
- School of Science, China Pharmaceutical University, 639 Longmian Avenue, Nanjing 211198, PR China
| | - Shilong Sun
- School of Science, China Pharmaceutical University, 639 Longmian Avenue, Nanjing 211198, PR China
| | - Tao Lu
- School of Science, China Pharmaceutical University, 639 Longmian Avenue, Nanjing 211198, PR China; State Key Laboratory of Natural Medicines, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing 210009, PR China
| | - Weifang Tang
- School of Science, China Pharmaceutical University, 639 Longmian Avenue, Nanjing 211198, PR China.
| | - Yong Zhu
- School of Science, China Pharmaceutical University, 639 Longmian Avenue, Nanjing 211198, PR China.
| |
Collapse
|
22
|
DIBAJI HASSAN, SHOTORBAN ALIKAZEMINASABAN, HABIBI MAHSA, GRATTAN RACHELM, LUCERO SHAYNA, SCHODT DAVIDJ, LIDKE KEITHA, PETRUCCELLI JONATHAN, LIDKE DIANES, LIU SHENG, CHAKRABORTY TONMOY. Axial de-scanning using remote focusing in the detection arm of light-sheet microscopy. RESEARCH SQUARE 2023:rs.3.rs-3338831. [PMID: 37886461 PMCID: PMC10602066 DOI: 10.21203/rs.3.rs-3338831/v1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/28/2023]
Abstract
The ability to image at high speeds is necessary in biological imaging to capture fast-moving or transient events or to efficiently image large samples. However, due to the lack of rigidity of biological specimens, carrying out fast, high-resolution volumetric imaging without moving and agitating the sample has been a challenging problem. Pupil-matched remote focusing has been promising for high NA imaging systems with their low aberrations and wavelength independence, making it suitable for multicolor imaging. However, owing to the incoherent and unpolarized nature of the fluorescence signal, manipulating this emission light through remote focusing is challenging. Therefore, remote focusing has been primarily limited to the illumination arm, using polarized laser light for facilitating coupling in and out of the remote focusing optics. Here we introduce a novel optical design that can de-scan the axial focus movement in the detection arm of a microscope. Our method splits the fluorescence signal into S and P-polarized light and lets them pass through the remote focusing module separately and combines them with the camera. This allows us to use only one focusing element to perform aberration-free, multi-color, volumetric imaging without (a) compromising the fluorescent signal and (b) needing to perform sample/detection-objective translation. We demonstrate the capabilities of this scheme by acquiring fast dual-color 4D (3D space + time) image stacks, with an axial range of 70 μm and camera limited acquisition speed. Owing to its general nature, we believe this technique will find its application to many other microscopy techniques that currently use an adjustable Z-stage to carry out volumetric imaging such as confocal, 2-photon, and light sheet variants.
Collapse
Affiliation(s)
- HASSAN DIBAJI
- Department of Physics and Astronomy, University of New Mexico, Albuquerque, New Mexico 87131, USA
| | | | - MAHSA HABIBI
- Department of Physics and Astronomy, University of New Mexico, Albuquerque, New Mexico 87131, USA
| | - RACHEL M GRATTAN
- Comprehensive Cancer Center, University of New Mexico Health Sciences Center, Albuquerque, New Mexico, 87131, USA
- Department of Pathology, University of New Mexico Health Science Center, Albuquerque, NM, USA
| | - SHAYNA LUCERO
- Comprehensive Cancer Center, University of New Mexico Health Sciences Center, Albuquerque, New Mexico, 87131, USA
- Department of Pathology, University of New Mexico Health Science Center, Albuquerque, NM, USA
| | - DAVID J. SCHODT
- Department of Physics and Astronomy, University of New Mexico, Albuquerque, New Mexico 87131, USA
| | - KEITH A. LIDKE
- Department of Physics and Astronomy, University of New Mexico, Albuquerque, New Mexico 87131, USA
- Comprehensive Cancer Center, University of New Mexico Health Sciences Center, Albuquerque, New Mexico, 87131, USA
| | - JONATHAN PETRUCCELLI
- Department of Physics, University at Albany–State University of NewYork,1400 Washington Avenue, Albany, NY 12222, USA
| | - DIANE S. LIDKE
- Comprehensive Cancer Center, University of New Mexico Health Sciences Center, Albuquerque, New Mexico, 87131, USA
- Department of Pathology, University of New Mexico Health Science Center, Albuquerque, NM, USA
| | - SHENG LIU
- Department of Physics and Astronomy, University of New Mexico, Albuquerque, New Mexico 87131, USA
| | - TONMOY CHAKRABORTY
- Department of Physics and Astronomy, University of New Mexico, Albuquerque, New Mexico 87131, USA
- Comprehensive Cancer Center, University of New Mexico Health Sciences Center, Albuquerque, New Mexico, 87131, USA
| |
Collapse
|
23
|
Liu R, Zhang Y, Wang Y, Huang Y, Gao J, Tian X, Ma T, Zhang T. Anti-inflammatory effect of dictamnine on allergic rhinitis via suppression of the LYN kinase-mediated molecular signaling pathway during mast cell activation. Phytother Res 2023; 37:4236-4250. [PMID: 37329155 DOI: 10.1002/ptr.7904] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2022] [Revised: 04/14/2023] [Accepted: 05/19/2023] [Indexed: 06/18/2023]
Abstract
Mast cells (MCs) are important therapeutic targets for allergic diseases. High-affinity immunoglobulin E (IgE) Fc receptors (FcεRI) trigger abnormal activation of MCs. Allergic rhinitis (AR) is an IgE-mediated antigen inhalation reaction that occurs in the nasal mucosa. MC aggravation and dysfunction were observed during the early stages of AR pathogenesis. Herb-derived dictamnine exhibits anti-inflammatory effects. Here, we investigated the pharmacological effects of herb-derived dictamnine on IgE-induced activation of MCs and an ovalbumin (OVA)-induced murine AR model. The results indicated that dictamnine attenuated OVA-induced local allergic reactions and reduced body temperature in OVA-challenged mice with active systemic anaphylaxis. Additionally, dictamnine decreased the frequency of nasal rubbing and sneezing in an OVA-induced murine AR model. Moreover, dictamnine inhibited FcεRI-activated MC activation in a dose-dependent manner without causing cytotoxicity, reduced the activation of the tyrosine kinase LYN in LAD2 cells, and downregulated the phosphorylation of PLCγ1, IP3R, PKC, Erk1/2, and Akt, which are downstream of LYN. In conclusion, dictamnine suppressed the OVA-stimulated murine model of AR and activated IgE-induced MCs via the LYN kinase-mediated molecular signaling pathway, suggesting that dictamnine may be a promising treatment for AR.
Collapse
Affiliation(s)
- Rui Liu
- School of Pharmacy, Xi'an Jiaotong University, Xi'an, China
| | - Yonghui Zhang
- School of Pharmacy, Xi'an Jiaotong University, Xi'an, China
| | - Yuejin Wang
- School of Pharmacy, Xi'an Jiaotong University, Xi'an, China
| | - Yihan Huang
- School of Pharmacy, Xi'an Jiaotong University, Xi'an, China
| | - Jiapan Gao
- School of Pharmacy, Xi'an Jiaotong University, Xi'an, China
| | - Xi Tian
- Department of Nephrology, The Affiliated Hospital of Northwest University, Xi'an, China
| | - Tianyou Ma
- School of Public Health, Xi'an Jiaotong University Health Science Center, Xi'an, China
| | - Tao Zhang
- School of Pharmacy, Xi'an Jiaotong University, Xi'an, China
| |
Collapse
|
24
|
Kanagaratham C, Derakhshan T, El Ansari YS, Furiness KN, Hollers E, Keldsen M, Oettgen HC, Dwyer DF. IgG:FcγRIIb signals block effector programs of IgE:FcεRI-activated mast cells but spare survival pathways. J Allergy Clin Immunol 2023; 152:453-468. [PMID: 37030590 PMCID: PMC10524869 DOI: 10.1016/j.jaci.2023.03.027] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Revised: 03/28/2023] [Accepted: 03/29/2023] [Indexed: 04/10/2023]
Abstract
BACKGROUND IgE-induced mast cell (MC) degranulation can be inhibited by IgG antibodies, signaling via FcγRIIb, but the effects of IgG on IgE-induced MC transcription are unknown. OBJECTIVE We sought to assess inhibitory IgG:FcγRIIb effects on MC responses to IgE using complementary transcriptomic and functional approaches. METHODS RNA sequencing was performed on bone marrow-derived MCs from wild-type and FcγRIIb-deficient mice to identify genes activated following IgE receptor crosslinking that were further modulated in the presence of antigen-specific IgG in an FcγRIIb-dependent fashion. Parallel analyses of signaling pathways and allergic responses in vivo were performed to assess the impact of these changes in gene expression. RESULTS Rapid changes in the transcription of 879 genes occurred in MCs activated by IgE, peaking at 1 hour. Surprisingly, only 12% of these were altered by IgG signaling via FcγRIIb, including numerous transcripts involved in orchestrating type 2 responses linked to spleen tyrosine kinase signaling. Consistent with this finding, IgG suppressed IgE-induced phospho-intermediates in the spleen tyrosine kinase signaling pathway. In vivo studies confirmed that the IgG-mediated suppression of both systemic anaphylaxis and MC-driven tissue recruitment of inflammatory cells following allergen challenge was dependent on FcγRIIb. In contrast, genes in the STAT5a cell survival pathway were unaltered by IgG, and STAT5a phosphorylation increased after IgE-induced MC activation but was unaffected by IgG. CONCLUSIONS Our findings indicate that inhibitory IgG:FcγRIIb signals block an IgE-induced proallergic program but spare a prosurvival program.
Collapse
Affiliation(s)
- Cynthia Kanagaratham
- Department of Pediatrics, Boston Children's Hospital, Boston, Mass; Department of Pediatrics, Harvard Medical School, Boston, Mass
| | - Tahereh Derakhshan
- Division of Allergy and Clinical Immunology, Jeff and Penny Vinik Center for Allergic Disease Research, Brigham and Women's Hospital, Boston, Mass; Department of Medicine, Harvard Medical School, Boston, Mass
| | - Yasmeen S El Ansari
- Department of Pediatrics, Boston Children's Hospital, Boston, Mass; Institute of Laboratory Medicine, Philipps University Marburg, Marburg, Germany
| | | | - Eleanor Hollers
- Division of Allergy and Clinical Immunology, Jeff and Penny Vinik Center for Allergic Disease Research, Brigham and Women's Hospital, Boston, Mass
| | - Mats Keldsen
- Department of Pediatrics, Boston Children's Hospital, Boston, Mass
| | - Hans C Oettgen
- Department of Pediatrics, Boston Children's Hospital, Boston, Mass; Department of Pediatrics, Harvard Medical School, Boston, Mass.
| | - Daniel F Dwyer
- Division of Allergy and Clinical Immunology, Jeff and Penny Vinik Center for Allergic Disease Research, Brigham and Women's Hospital, Boston, Mass; Department of Medicine, Harvard Medical School, Boston, Mass.
| |
Collapse
|
25
|
Bashore FM, Katis VL, Du Y, Sikdar A, Wang D, Bradshaw WJ, Rygiel KA, Leisner TM, Chalk R, Mishra S, Williams AC, Gileadi O, Brennan PE, Wiley JC, Gockley J, Cary GA, Carter GW, Young JE, Pearce KH, Fu H, Axtman AD. Characterization of covalent inhibitors that disrupt the interaction between the tandem SH2 domains of SYK and FCER1G phospho-ITAM. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.07.28.551026. [PMID: 37547005 PMCID: PMC10402180 DOI: 10.1101/2023.07.28.551026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/08/2023]
Abstract
RNA sequencing and genetic data support spleen tyrosine kinase (SYK) and high affinity immunoglobulin epsilon receptor subunit gamma (FCER1G) as putative targets to be modulated for Alzheimer's disease (AD) therapy. FCER1G is a component of Fc receptor complexes that contain an immunoreceptor tyrosine-based activation motif (ITAM). SYK interacts with the Fc receptor by binding to doubly phosphorylated ITAM (p-ITAM) via its two tandem SH2 domains (SYK-tSH2). Interaction of the FCER1G p-ITAM with SYK-tSH2 enables SYK activation via phosphorylation. Since SYK activation is reported to exacerbate AD pathology, we hypothesized that disruption of this interaction would be beneficial for AD patients. Herein, we developed biochemical and biophysical assays to enable the discovery of small molecules that perturb the interaction between the FCER1G p-ITAM and SYK-tSH2. We identified two distinct chemotypes using a high-throughput screen (HTS) and orthogonally assessed their binding. Both chemotypes covalently modify SYK-tSH2 and inhibit its interaction with FCER1G p-ITAM.
Collapse
Affiliation(s)
- Frances M Bashore
- UNC Eshelman School of Pharmacy, Division of Chemical Biology and Medicinal Chemistry, Structural Genomics Consortium, University of North Carolina, Chapel Hill, NC, USA
| | - Vittorio L Katis
- ARUK Oxford Drug Discovery Institute, Centre for Medicines Discovery, Nuffield Department of Medicine Research Building, Old Road Campus, University of Oxford, Headington, Oxford, OX3 7FZ, UK
| | - Yuhong Du
- Department of Pharmacology and Chemical Biology, School of Medicine, Emory University, Atlanta, GA, USA; Emory Chemical Biology Discovery Center, School of Medicine, Emory University, Atlanta, GA, USA
| | - Arunima Sikdar
- UNC Eshelman School of Pharmacy, Division of Chemical Biology and Medicinal Chemistry, Center for Integrative Chemical Biology and Drug Discovery, University of North Carolina, Chapel Hill, NC, USA
| | - Dongxue Wang
- Department of Pharmacology and Chemical Biology, School of Medicine, Emory University, Atlanta, GA, USA; Emory Chemical Biology Discovery Center, School of Medicine, Emory University, Atlanta, GA, USA
| | - William J Bradshaw
- ARUK Oxford Drug Discovery Institute, Centre for Medicines Discovery, Nuffield Department of Medicine Research Building, Old Road Campus, University of Oxford, Headington, Oxford, OX3 7FZ, UK
| | - Karolina A Rygiel
- ARUK Oxford Drug Discovery Institute, Centre for Medicines Discovery, Nuffield Department of Medicine Research Building, Old Road Campus, University of Oxford, Headington, Oxford, OX3 7FZ, UK
| | - Tina M Leisner
- UNC Eshelman School of Pharmacy, Division of Chemical Biology and Medicinal Chemistry, Center for Integrative Chemical Biology and Drug Discovery, University of North Carolina, Chapel Hill, NC, USA
| | - Rod Chalk
- ARUK Oxford Drug Discovery Institute, Centre for Medicines Discovery, Nuffield Department of Medicine Research Building, Old Road Campus, University of Oxford, Headington, Oxford, OX3 7FZ, UK
| | | | | | - Opher Gileadi
- ARUK Oxford Drug Discovery Institute, Centre for Medicines Discovery, Nuffield Department of Medicine Research Building, Old Road Campus, University of Oxford, Headington, Oxford, OX3 7FZ, UK
- Current address: Structural Genomics Consortium, Department of Medicine, Karolinska Hospital and Karolinska Institute, 171 76 Stockholm, Sweden
| | - Paul E Brennan
- ARUK Oxford Drug Discovery Institute, Centre for Medicines Discovery, Nuffield Department of Medicine Research Building, Old Road Campus, University of Oxford, Headington, Oxford, OX3 7FZ, UK
| | | | | | | | | | | | - Kenneth H Pearce
- UNC Eshelman School of Pharmacy, Division of Chemical Biology and Medicinal Chemistry, Center for Integrative Chemical Biology and Drug Discovery, University of North Carolina, Chapel Hill, NC, USA
| | - Haian Fu
- Department of Pharmacology and Chemical Biology, School of Medicine, Emory University, Atlanta, GA, USA; Emory Chemical Biology Discovery Center, School of Medicine, Emory University, Atlanta, GA, USA
| | - Alison D Axtman
- UNC Eshelman School of Pharmacy, Division of Chemical Biology and Medicinal Chemistry, Structural Genomics Consortium, University of North Carolina, Chapel Hill, NC, USA
| |
Collapse
|
26
|
Choi YA, Dhakal H, Lee S, Kim N, Lee B, Kwon TK, Khang D, Kim SH. IRF3 Activation in Mast Cells Promotes FcεRI-Mediated Allergic Inflammation. Cells 2023; 12:1493. [PMID: 37296614 PMCID: PMC10252328 DOI: 10.3390/cells12111493] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Revised: 05/22/2023] [Accepted: 05/26/2023] [Indexed: 06/12/2023] Open
Abstract
(1) Background: This study aims to elucidate a novel non-transcriptional action of IRF3 in addition to its role as a transcription factor in mast cell activation and associated allergic inflammation; (2) Methods: For in vitro experiments, mouse bone-marrow-derived mast cells (mBMMCs) and a rat basophilic leukemia cell line (RBL-2H3) were used for investigating the underlying mechanism of IRF3 in mast-cell-mediated allergic inflammation. For in vivo experiments, wild-type and Irf3 knockout mice were used for evaluating IgE-mediated local and systemic anaphylaxis; (3) Results: Passive cutaneous anaphylaxis (PCA)-induced tissues showed highly increased IRF3 activity. In addition, the activation of IRF3 was observed in DNP-HSA-treated mast cells. Phosphorylated IRF3 by DNP-HSA was spatially co-localized with tryptase according to the mast cell activation process, and FcεRI-mediated signaling pathways directly regulated that activity. The alteration of IRF3 affected the production of granule contents in the mast cells and the anaphylaxis responses, including PCA- and ovalbumin-induced active systemic anaphylaxis. Furthermore, IRF3 influenced the post-translational processing of histidine decarboxylase (HDC), which is required for granule maturation; and (4) Conclusion: Through this study, we demonstrated the novel function of IRF3 as an important factor inducing mast cell activation and as an upstream molecule for HDC activity.
Collapse
Affiliation(s)
- Young-Ae Choi
- Cell & Matrix Research Institute, Department of Pharmacology, School of Medicine, Kyungpook National University, Daegu 41944, Republic of Korea; (Y.-A.C.); (H.D.); (N.K.)
| | - Hima Dhakal
- Cell & Matrix Research Institute, Department of Pharmacology, School of Medicine, Kyungpook National University, Daegu 41944, Republic of Korea; (Y.-A.C.); (H.D.); (N.K.)
| | - Soyoung Lee
- Immunoregulatory Materials Research Center, Korea Research Institute of Bioscience and Biotechnology, Jeongeup 56212, Republic of Korea;
| | - Namkyung Kim
- Cell & Matrix Research Institute, Department of Pharmacology, School of Medicine, Kyungpook National University, Daegu 41944, Republic of Korea; (Y.-A.C.); (H.D.); (N.K.)
| | - Byungheon Lee
- Department of Biochemistry and Cell Biology, School of Medicine, Kyungpook National University, Daegu 41944, Republic of Korea;
| | - Taeg Kyu Kwon
- Department of Immunology, School of Medicine, Keimyung University, Daegu 42601, Republic of Korea;
| | - Dongwoo Khang
- Department of Physiology, School of Medicine, Gachon University, Incheon 21999, Republic of Korea
| | - Sang-Hyun Kim
- Cell & Matrix Research Institute, Department of Pharmacology, School of Medicine, Kyungpook National University, Daegu 41944, Republic of Korea; (Y.-A.C.); (H.D.); (N.K.)
| |
Collapse
|
27
|
Chang D, Hammer C, Holweg CTJ, Selvaraj S, Rathore N, McCarthy MI, Yaspan BL, Choy DF. A genome-wide association study of chronic spontaneous urticaria risk and heterogeneity. J Allergy Clin Immunol 2023; 151:1351-1356. [PMID: 36343773 DOI: 10.1016/j.jaci.2022.10.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Revised: 10/10/2022] [Accepted: 10/14/2022] [Indexed: 11/06/2022]
Abstract
BACKGROUND Chronic spontaneous urticaria (CSU) is a dermatologic condition characterized by spontaneous, pruritic hives and/or angioedema that persists for 6 weeks or longer with no identifiable trigger. Antihistamines and second-line therapies such as omalizumab are effective for some CSU patients, but others remain symptomatic, with significant impact on quality of life. This variable response to treatment and autoantibody levels across patients highlight clinically heterogeneous subgroups. OBJECTIVE We aimed to highlight pathways involved in CSU by investigating the genetics of CSU risk and subgroups. METHODS We performed a genome-wide association study (GWAS) of 679 CSU patients and 4446 controls and a GWAS of chronic urticaria (CU)-index, which measures IgG autoantibodies levels, by comparing 447 CU index-low to 183 CU index-high patients. We also tested whether polygenic scores for autoimmune-related disorders were associated with CSU risk and CU index. RESULTS We identified 2 loci significantly associated with disease risk. The strongest association mapped to position 56 of HLA-DQA1 (P = 1.69 × 10-9), where the arginine residue was associated with increased risk (odds ratio = 1.64). The second association signal colocalized with expression-quantitative trait loci for ITPKB in whole blood (Pcolocalization = .997). The arginine residue at position 56 of HLA-DQA1 was also associated with increased risk of CU index-high (P = 6.15 × 10-5, odds ratio = 1.86), while the ITKPB association was not (P = .64). Polygenic scores for 3 autoimmune-related disorders (hypothyroidism, type 1 diabetes, and vitiligo) were associated with CSU risk and CU index (P < 2.34 × 10-3, odds ratio > 1.72). CONCLUSION A GWAS of CSU identified 2 genome-wide significant loci, highlighting the shared genetics between CU index and autoimmune disorders.
Collapse
Affiliation(s)
- Diana Chang
- Department of Human Genetics, Genentech Inc, South San Francisco, Calif.
| | - Christian Hammer
- Department of Human Genetics, Genentech Inc, South San Francisco, Calif; Department of Cancer Immunology, Genentech Inc, South San Francisco, Calif
| | | | - Suresh Selvaraj
- Department of Biosample and Repository Management, Genentech Inc, South San Francisco, Calif
| | - Nisha Rathore
- Biomarker Discovery OMNI, Genentech Inc, South San Francisco, Calif
| | - Mark I McCarthy
- Department of Human Genetics, Genentech Inc, South San Francisco, Calif
| | - Brian L Yaspan
- Department of Human Genetics, Genentech Inc, South San Francisco, Calif
| | - David F Choy
- Biomarker Discovery OMNI, Genentech Inc, South San Francisco, Calif
| |
Collapse
|
28
|
Wise SK, Damask C, Roland LT, Ebert C, Levy JM, Lin S, Luong A, Rodriguez K, Sedaghat AR, Toskala E, Villwock J, Abdullah B, Akdis C, Alt JA, Ansotegui IJ, Azar A, Baroody F, Benninger MS, Bernstein J, Brook C, Campbell R, Casale T, Chaaban MR, Chew FT, Chambliss J, Cianferoni A, Custovic A, Davis EM, DelGaudio JM, Ellis AK, Flanagan C, Fokkens WJ, Franzese C, Greenhawt M, Gill A, Halderman A, Hohlfeld JM, Incorvaia C, Joe SA, Joshi S, Kuruvilla ME, Kim J, Klein AM, Krouse HJ, Kuan EC, Lang D, Larenas-Linnemann D, Laury AM, Lechner M, Lee SE, Lee VS, Loftus P, Marcus S, Marzouk H, Mattos J, McCoul E, Melen E, Mims JW, Mullol J, Nayak JV, Oppenheimer J, Orlandi RR, Phillips K, Platt M, Ramanathan M, Raymond M, Rhee CS, Reitsma S, Ryan M, Sastre J, Schlosser RJ, Schuman TA, Shaker MS, Sheikh A, Smith KA, Soyka MB, Takashima M, Tang M, Tantilipikorn P, Taw MB, Tversky J, Tyler MA, Veling MC, Wallace D, Wang DY, White A, Zhang L. International consensus statement on allergy and rhinology: Allergic rhinitis - 2023. Int Forum Allergy Rhinol 2023; 13:293-859. [PMID: 36878860 DOI: 10.1002/alr.23090] [Citation(s) in RCA: 160] [Impact Index Per Article: 80.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Revised: 11/11/2022] [Accepted: 09/13/2022] [Indexed: 03/08/2023]
Abstract
BACKGROUND In the 5 years that have passed since the publication of the 2018 International Consensus Statement on Allergy and Rhinology: Allergic Rhinitis (ICAR-Allergic Rhinitis 2018), the literature has expanded substantially. The ICAR-Allergic Rhinitis 2023 update presents 144 individual topics on allergic rhinitis (AR), expanded by over 40 topics from the 2018 document. Originally presented topics from 2018 have also been reviewed and updated. The executive summary highlights key evidence-based findings and recommendation from the full document. METHODS ICAR-Allergic Rhinitis 2023 employed established evidence-based review with recommendation (EBRR) methodology to individually evaluate each topic. Stepwise iterative peer review and consensus was performed for each topic. The final document was then collated and includes the results of this work. RESULTS ICAR-Allergic Rhinitis 2023 includes 10 major content areas and 144 individual topics related to AR. For a substantial proportion of topics included, an aggregate grade of evidence is presented, which is determined by collating the levels of evidence for each available study identified in the literature. For topics in which a diagnostic or therapeutic intervention is considered, a recommendation summary is presented, which considers the aggregate grade of evidence, benefit, harm, and cost. CONCLUSION The ICAR-Allergic Rhinitis 2023 update provides a comprehensive evaluation of AR and the currently available evidence. It is this evidence that contributes to our current knowledge base and recommendations for patient evaluation and treatment.
Collapse
Affiliation(s)
- Sarah K Wise
- Otolaryngology-HNS, Emory University, Atlanta, Georgia, USA
| | - Cecelia Damask
- Otolaryngology-HNS, Private Practice, University of Central Florida, Lake Mary, Florida, USA
| | - Lauren T Roland
- Otolaryngology-HNS, Washington University, St. Louis, Missouri, USA
| | - Charles Ebert
- Otolaryngology-HNS, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Joshua M Levy
- Otolaryngology-HNS, Emory University, Atlanta, Georgia, USA
| | - Sandra Lin
- Otolaryngology-HNS, University of Wisconsin, Madison, Wisconsin, USA
| | - Amber Luong
- Otolaryngology-HNS, McGovern Medical School of the University of Texas, Houston, Texas, USA
| | - Kenneth Rodriguez
- Otolaryngology-HNS, University Hospitals Cleveland Medical Center, Cleveland, Ohio, USA
| | - Ahmad R Sedaghat
- Otolaryngology-HNS, University of Cincinnati, Cincinnati, Ohio, USA
| | - Elina Toskala
- Otolaryngology-HNS, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | | | - Baharudin Abdullah
- Otolaryngology-HNS, Universiti Sains Malaysia, Kubang, Kerian, Kelantan, Malaysia
| | - Cezmi Akdis
- Immunology, Infectious Diseases, Swiss Institute of Allergy and Asthma Research, Davos, Switzerland
| | - Jeremiah A Alt
- Otolaryngology-HNS, University of Utah, Salt Lake City, Utah, USA
| | | | - Antoine Azar
- Allergy/Immunology, Johns Hopkins University, Baltimore, Maryland, USA
| | - Fuad Baroody
- Otolaryngology-HNS, University of Chicago, Chicago, Illinois, USA
| | | | | | - Christopher Brook
- Otolaryngology-HNS, Harvard University, Beth Israel Deaconess Medical Center, Boston, Massachusetts, USA
| | - Raewyn Campbell
- Otolaryngology-HNS, Macquarie University, Sydney, NSW, Australia
| | - Thomas Casale
- Allergy/Immunology, University of South Florida College of Medicine, Tampa, Florida, USA
| | - Mohamad R Chaaban
- Otolaryngology-HNS, Cleveland Clinic, Case Western Reserve University, Cleveland, Ohio, USA
| | - Fook Tim Chew
- Allergy/Immunology, Genetics, National University of Singapore, Singapore, Singapore
| | - Jeffrey Chambliss
- Allergy/Immunology, University of Texas Southwestern, Dallas, Texas, USA
| | - Antonella Cianferoni
- Allergy/Immunology, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | | | | | | | - Anne K Ellis
- Allergy/Immunology, Queens University, Kingston, ON, Canada
| | | | - Wytske J Fokkens
- Otorhinolaryngology, Amsterdam University Medical Centres, Amsterdam, Netherlands
| | | | - Matthew Greenhawt
- Allergy/Immunology, Pediatrics, University of Colorado, Children's Hospital Colorado, Aurora, Colorado, USA
| | - Amarbir Gill
- Otolaryngology-HNS, University of Michigan, Ann Arbor, Michigan, USA
| | - Ashleigh Halderman
- Otolaryngology-HNS, University of Texas Southwestern, Dallas, Texas, USA
| | - Jens M Hohlfeld
- Respiratory Medicine, Fraunhofer Institute for Toxicology and Experimental Medicine ITEM, Hannover Medical School, German Center for Lung Research, Hannover, Germany
| | | | - Stephanie A Joe
- Otolaryngology-HNS, University of Illinois Chicago, Chicago, Illinois, USA
| | - Shyam Joshi
- Allergy/Immunology, Oregon Health and Science University, Portland, Oregon, USA
| | | | - Jean Kim
- Otolaryngology-HNS, Johns Hopkins University, Baltimore, Maryland, USA
| | - Adam M Klein
- Otolaryngology-HNS, Emory University, Atlanta, Georgia, USA
| | - Helene J Krouse
- Otorhinolaryngology Nursing, University of Texas Rio Grande Valley, Edinburg, Texas, USA
| | - Edward C Kuan
- Otolaryngology-HNS, University of California Irvine, Orange, California, USA
| | - David Lang
- Allergy/Immunology, Cleveland Clinic, Cleveland, Ohio, USA
| | | | | | - Matt Lechner
- Otolaryngology-HNS, University College London, Barts Health NHS Trust, London, UK
| | - Stella E Lee
- Otolaryngology-HNS, Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - Victoria S Lee
- Otolaryngology-HNS, University of Illinois Chicago, Chicago, Illinois, USA
| | - Patricia Loftus
- Otolaryngology-HNS, University of California San Francisco, San Francisco, California, USA
| | - Sonya Marcus
- Otolaryngology-HNS, Stony Brook University, Stony Brook, New York, USA
| | - Haidy Marzouk
- Otolaryngology-HNS, State University of New York Upstate, Syracuse, New York, USA
| | - Jose Mattos
- Otolaryngology-HNS, University of Virginia, Charlottesville, Virginia, USA
| | - Edward McCoul
- Otolaryngology-HNS, Ochsner Clinic, New Orleans, Louisiana, USA
| | - Erik Melen
- Pediatric Allergy, Karolinska Institutet, Stockholm, Sweden
| | - James W Mims
- Otolaryngology-HNS, Wake Forest University, Winston Salem, North Carolina, USA
| | - Joaquim Mullol
- Otorhinolaryngology, Hospital Clinic Barcelona, Barcelona, Spain
| | - Jayakar V Nayak
- Otolaryngology-HNS, Stanford University, Palo Alto, California, USA
| | - John Oppenheimer
- Allergy/Immunology, Rutgers, State University of New Jersey, Newark, New Jersey, USA
| | | | - Katie Phillips
- Otolaryngology-HNS, University of Cincinnati, Cincinnati, Ohio, USA
| | - Michael Platt
- Otolaryngology-HNS, Boston University, Boston, Massachusetts, USA
| | | | | | - Chae-Seo Rhee
- Rhinology/Allergy, Seoul National University Hospital and College of Medicine, Seoul, Korea
| | - Sietze Reitsma
- Otolaryngology-HNS, University of Amsterdam, Amsterdam, Netherlands
| | - Matthew Ryan
- Otolaryngology-HNS, University of Texas Southwestern, Dallas, Texas, USA
| | - Joaquin Sastre
- Allergy, Fundacion Jiminez Diaz, University Autonoma de Madrid, Madrid, Spain
| | - Rodney J Schlosser
- Otolaryngology-HNS, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Theodore A Schuman
- Otolaryngology-HNS, Virginia Commonwealth University, Richmond, Virginia, USA
| | - Marcus S Shaker
- Allergy/Immunology, Dartmouth Geisel School of Medicine, Lebanon, New Hampshire, USA
| | - Aziz Sheikh
- Primary Care, University of Edinburgh, Edinburgh, Scotland
| | - Kristine A Smith
- Otolaryngology-HNS, University of Utah, Salt Lake City, Utah, USA
| | - Michael B Soyka
- Otolaryngology-HNS, University of Zurich, University Hospital of Zurich, Zurich, Switzerland
| | - Masayoshi Takashima
- Otolaryngology-HNS, Houston Methodist Academic Institute, Houston, Texas, USA
| | - Monica Tang
- Allergy/Immunology, University of California San Francisco, San Francisco, California, USA
| | | | - Malcolm B Taw
- Integrative East-West Medicine, University of California Los Angeles, Westlake Village, California, USA
| | - Jody Tversky
- Allergy/Immunology, Johns Hopkins University, Baltimore, Maryland, USA
| | - Matthew A Tyler
- Otolaryngology-HNS, University of Minnesota, Minneapolis, Minnesota, USA
| | - Maria C Veling
- Otolaryngology-HNS, University of Texas Southwestern, Dallas, Texas, USA
| | - Dana Wallace
- Allergy/Immunology, Nova Southeastern University, Ft. Lauderdale, Florida, USA
| | - De Yun Wang
- Otolaryngology-HNS, National University of Singapore, Singapore, Singapore
| | - Andrew White
- Allergy/Immunology, Scripps Clinic, San Diego, California, USA
| | - Luo Zhang
- Otolaryngology-HNS, Beijing Tongren Hospital, Beijing, China
| |
Collapse
|
29
|
Adnan A, Acharya S, Alenazy LA, de las Vecillas L, Giavina Bianchi P, Picard M, Calbache-Gil L, Romero-Pinedo S, Abadí´a-Molina AC, Kerr W, Pedicone C, Nagai J, Hollers E, Dwyer D, Castells M. Multistep IgE Mast Cell Desensitization Is a Dose- and Time-Dependent Process Partially Regulated by SHIP-1. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2023; 210:709-720. [PMID: 36881903 PMCID: PMC9986054 DOI: 10.4049/jimmunol.2100485] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Accepted: 12/22/2022] [Indexed: 03/09/2023]
Abstract
Multistep mast cell desensitization blocks the release of mediators following IgE crosslinking with increasing doses of Ag. Although its in vivo application has led to the safe reintroduction of drugs and foods in IgE-sensitized patients at risk for anaphylaxis, the mechanisms of the inhibitory process have remained elusive. We sought to investigate the kinetics, membrane, and cytoskeletal changes and to identify molecular targets. IgE-sensitized wild-type murine (WT) and FcεRIα humanized (h) bone marrow mast cells were activated and desensitized with DNP, nitrophenyl, dust mites, and peanut Ags. The movements of membrane receptors, FcεRI/IgE/Ag, actin, and tubulin and the phosphorylation of Syk, Lyn, P38-MAPK, and SHIP-1 were assessed. Silencing SHIP-1 protein was used to dissect the SHIP-1 role. Multistep IgE desensitization of WT and transgenic human bone marrow mast cells blocked the release of β-hexosaminidase in an Ag-specific fashion and prevented actin and tubulin movements. Desensitization was regulated by the initial Ag dose, number of doses, and time between doses. FcεRI, IgE, Ags, and surface receptors were not internalized during desensitization. Phosphorylation of Syk, Lyn, p38 MAPK, and SHIP-1 increased in a dose-response manner during activation; in contrast, only SHIP-1 phosphorylation increased in early desensitization. SHIP-1 phosphatase function had no impact on desensitization, but silencing SHIP-1 increased β-hexoxaminidase release, preventing desensitization. Multistep IgE mast cell desensitization is a dose- and time-regulated process that blocks β-hexosaminidase, impacting membrane and cytoskeletal movements. Signal transduction is uncoupled, favoring early phosphorylation of SHIP-1. Silencing SHIP-1 impairs desensitization without implicating its phosphatase function.
Collapse
Affiliation(s)
- Ather Adnan
- Division of Allergy and Immunology, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
- Texas A&M Health Science Center, College of Medicine, Houston, TX
| | - Shree Acharya
- Division of Allergy and Immunology, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
| | - Leila A. Alenazy
- Division of Allergy and Immunology, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
- Division of Allergy and Clinical Immunology, Department of Medicine, College of Medicine, King Saud University, Riyadh, Saudi Arabia
| | - Leticia de las Vecillas
- Division of Allergy and Immunology, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
- Department of Allergy, Marqués de Valdecilla University Hospital – Instituto de Investigación Marques de Valdecilla, Santander, Spain
| | - Pedro Giavina Bianchi
- Clinical Immunology and Allergy Division, School of Medicine, Universidade de São Paulo, R. Prof. Artur Ramos Sao Paulo, Brazil
| | - Matthieu Picard
- Division of Allergy and Clinical Immunology, Department of Medicine, Hôpital Maisonneuve-Rosemont, Université de Montréal, Montreal, QC, Canada
| | - Lucia Calbache-Gil
- Unidad de Inmunología, IBIMER, CIBM, Universidad de Granada, Granada, Spain
- Departamento de Bioquímica y Biología Molecular III e Inmunología, Facultad de Medicina, Universidad de Granada, Granada, Spain
| | - Salvador Romero-Pinedo
- Unidad de Inmunología, IBIMER, CIBM, Universidad de Granada, Granada, Spain
- Departamento de Bioquímica y Biología Molecular III e Inmunología, Facultad de Medicina, Universidad de Granada, Granada, Spain
| | - Ana Clara Abadí´a-Molina
- Unidad de Inmunología, IBIMER, CIBM, Universidad de Granada, Granada, Spain
- Departamento de Bioquímica y Biología Molecular III e Inmunología, Facultad de Medicina, Universidad de Granada, Granada, Spain
| | - William Kerr
- Department of Microbiology and Immunology, SUNY Upstate Medical University, Syracuse, NY
| | - Chiara Pedicone
- Department of Microbiology and Immunology, SUNY Upstate Medical University, Syracuse, NY
| | - Jun Nagai
- Division of Allergy and Immunology, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
| | - Eleanor Hollers
- Division of Allergy and Immunology, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
| | - Daniel Dwyer
- Division of Allergy and Immunology, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
| | - Mariana Castells
- Division of Allergy and Immunology, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
| |
Collapse
|
30
|
Lee JE, Choi MY, Min KY, Jo MG, Kim YM, Kim HS, Choi WS. Drug repositioning of anti-microbial agent nifuratel to treat mast cell-mediated allergic responses. Int J Immunopathol Pharmacol 2023; 37:3946320231202349. [PMID: 37706235 PMCID: PMC10503296 DOI: 10.1177/03946320231202349] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Accepted: 09/01/2023] [Indexed: 09/15/2023] Open
Abstract
Objectives: Our objective was to assess the effects and mechanisms of nifuratel on IgE-mediated mast cell (MC) degranulation and anaphylaxis in both in vitro and in vivo settings.Methods: The anti-allergic activity of nifuratel was evaluated in mast cell cultures and the passive cutaneous anaphylaxis (PCA) model. The effects of nifuratel on signaling pathways stimulated by antigen in mast cells were measured by immunoblotting, immunoprecipitation, in vitro protein tyrosine kinase assay, and other molecular biological methods.Results: Nifuratel reversibly inhibited antigen-induced degranulation of MCs (IC50, approximately 0.34 μM for RBL-2H3 cells; approximately 0.94 μM for BMMCs) and suppressed the secretion of inflammatory cytokines IL-4 (IC50, approximately 0.74 μM) and TNF-α (IC50, approximately 0.48 μM). Mechanism studies showed that nifuratel inhibited the phosphorylation of Syk by antigen via the inhibition of recruitment of cytosolic Syk to the ɣ subunit of FcεRI, and decreased the activation of Syk downstream signaling proteins LAT, Akt, and MAPKs. Finally, nifuratel dose-dependently suppressed the IgE-mediated passive cutaneous anaphylaxis in mice (ED50, approximately 22 mg/kg).Conclusion: Our findings suggest that nifuratel inhibits pathways essential for the activation of mast cells to suppress anaphylaxis, thereby indicating that the anti-microbial drug, nifuratel, could be a potential drug candidate for IgE-mediated allergic disorders.
Collapse
Affiliation(s)
- Ji Eon Lee
- Department of Immunology, College of Medicine, Konkuk University, Chungju, Republic of Korea
| | - Min Yeong Choi
- Department of Immunology, College of Medicine, Konkuk University, Chungju, Republic of Korea
| | - Keun Young Min
- Department of Immunology, College of Medicine, Konkuk University, Chungju, Republic of Korea
| | - Min Geun Jo
- Department of Immunology, College of Medicine, Konkuk University, Chungju, Republic of Korea
| | - Young Mi Kim
- College of Pharmacy, Duksung Women’s University, Seoul, Republic of Korea
| | - Hyuk Soon Kim
- Department of Biomedical Sciences, College of Natural Science and Department of Health Sciences, The Graduate School of Dong-A University, Busan, Republic of Korea
| | - Wahn Soo Choi
- Department of Immunology, College of Medicine, Konkuk University, Chungju, Republic of Korea
| |
Collapse
|
31
|
Bratti M, Vibhushan S, Longé C, Koumantou D, Ménasché G, Benhamou M, Varin-Blank N, Blank U, Saveanu L, Ben Mkaddem S. Insulin-regulated aminopeptidase contributes to setting the intensity of FcR-mediated inflammation. Front Immunol 2022; 13:1029759. [PMID: 36389775 PMCID: PMC9647545 DOI: 10.3389/fimmu.2022.1029759] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2022] [Accepted: 10/12/2022] [Indexed: 11/25/2022] Open
Abstract
The function of intracellular trafficking in immune-complex triggered inflammation remains poorly understood. Here, we investigated the role of Insulin-Regulated Amino Peptidase (IRAP)-positive endosomal compartments in Fc receptor (FcR)-induced inflammation. Less severe FcγR-triggered arthritis, active systemic anaphylaxis and FcεRI-triggered passive systemic anaphylaxis were observed in IRAP-deficient versus wild-type mice. In mast cells FcεRI stimulation induced rapid plasma membrane recruitment of IRAP-positive endosomes. IRAP-deficient cells exhibited reduced secretory responses, calcium signaling and activating SykY519/520 phosphorylation albeit receptor tyrosine phosphorylation on β and γ subunits was not different. By contrast, in the absence of IRAP, SHP1-inactivating phosphorylation on Ser591 that controls Syk activity was decreased. Ex-vivo cell profiling after FcγR-triggered anaphylaxis confirmed decreased phosphorylation of both SykY519/520 and SHP-1S591 in IRAP-deficient neutrophils and monocytes. Thus, IRAP-positive endosomal compartments, in promoting inhibition of SHP-1 during FcR signaling, control the extent of phosphorylation events at the plasma membrane and contribute to setting the intensity of immune-complex triggered inflammatory diseases.
Collapse
Affiliation(s)
- Manuela Bratti
- Université Paris Cité, Centre de Recherche sur l’Inflammation, Institut National de la Santé et de la Recherche Médicale (INSERM) Unité Mixte de Recherche (UMR)1149, Centre National de la Recherche Scientifique (CNRS) Equipe Mixte de Recherche(EMR)-8252, Faculté de Médecine site Bichat, Paris, France
- Université Paris Cité, Laboratoire d’Excellence INFLAMEX, Paris, France
| | - Shamila Vibhushan
- Université Paris Cité, Centre de Recherche sur l’Inflammation, Institut National de la Santé et de la Recherche Médicale (INSERM) Unité Mixte de Recherche (UMR)1149, Centre National de la Recherche Scientifique (CNRS) Equipe Mixte de Recherche(EMR)-8252, Faculté de Médecine site Bichat, Paris, France
- Université Paris Cité, Laboratoire d’Excellence INFLAMEX, Paris, France
| | - Cyril Longé
- Université Paris Cité, Imagine Institute, Laboratory of Molecular basis of altered immune homeostasis, Institut National de la Santé et de la Recherche Médicale (INSERM) Unité Mixte de Recherche (UMR)1163, Paris, France
| | - Despoina Koumantou
- Université Paris Cité, Centre de Recherche sur l’Inflammation, Institut National de la Santé et de la Recherche Médicale (INSERM) Unité Mixte de Recherche (UMR)1149, Centre National de la Recherche Scientifique (CNRS) Equipe Mixte de Recherche(EMR)-8252, Faculté de Médecine site Bichat, Paris, France
- Université Paris Cité, Laboratoire d’Excellence INFLAMEX, Paris, France
| | - Gaël Ménasché
- Université Paris Cité, Imagine Institute, Laboratory of Molecular basis of altered immune homeostasis, Institut National de la Santé et de la Recherche Médicale (INSERM) Unité Mixte de Recherche (UMR)1163, Paris, France
| | - Marc Benhamou
- Université Paris Cité, Centre de Recherche sur l’Inflammation, Institut National de la Santé et de la Recherche Médicale (INSERM) Unité Mixte de Recherche (UMR)1149, Centre National de la Recherche Scientifique (CNRS) Equipe Mixte de Recherche(EMR)-8252, Faculté de Médecine site Bichat, Paris, France
- Université Paris Cité, Laboratoire d’Excellence INFLAMEX, Paris, France
| | - Nadine Varin-Blank
- Institut National de la Santé et de la Recherche Médicale (INSERM) U978, Université Paris 13 Sorbonne Paris Nord, Unité de Formation et de Recherche (UFR) Santé Médecine et Biologie Humaine (SMBH), Bobigny, France
| | - Ulrich Blank
- Université Paris Cité, Centre de Recherche sur l’Inflammation, Institut National de la Santé et de la Recherche Médicale (INSERM) Unité Mixte de Recherche (UMR)1149, Centre National de la Recherche Scientifique (CNRS) Equipe Mixte de Recherche(EMR)-8252, Faculté de Médecine site Bichat, Paris, France
- Université Paris Cité, Laboratoire d’Excellence INFLAMEX, Paris, France
- *Correspondence: Ulrich Blank,
| | - Loredana Saveanu
- Université Paris Cité, Centre de Recherche sur l’Inflammation, Institut National de la Santé et de la Recherche Médicale (INSERM) Unité Mixte de Recherche (UMR)1149, Centre National de la Recherche Scientifique (CNRS) Equipe Mixte de Recherche(EMR)-8252, Faculté de Médecine site Bichat, Paris, France
- Université Paris Cité, Laboratoire d’Excellence INFLAMEX, Paris, France
| | - Sanae Ben Mkaddem
- Institut National de la Santé et de la Recherche Médicale (INSERM) U978, Université Paris 13 Sorbonne Paris Nord, Unité de Formation et de Recherche (UFR) Santé Médecine et Biologie Humaine (SMBH), Bobigny, France
- Institute of biological Sciences, Mohammed VI Polytechnic University (UM6P), Ben-Guerir, Morocco
| |
Collapse
|
32
|
The Role of TGFβ and Other Cytokines in Regulating Mast Cell Functions in Allergic Inflammation. Int J Mol Sci 2022; 23:ijms231810864. [PMID: 36142776 PMCID: PMC9503477 DOI: 10.3390/ijms231810864] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Revised: 09/12/2022] [Accepted: 09/15/2022] [Indexed: 12/15/2022] Open
Abstract
Mast cells (MC) are a key effector cell in multiple types of immune responses, including atopic conditions. Allergic diseases have been steadily rising across the globe, creating a growing public health problem. IgE-mediated activation of MCs leads to the release of potent mediators that can have dire clinical consequences. Current therapeutic options to inhibit MC activation and degranulation are limited; thus, a better understanding of the mechanisms that regulate MC effector functions in allergic inflammation are necessary in order to develop effective treatment options with minimal side effects. Several cytokines have been identified that play multifaceted roles in regulating MC activation, including TGFβ, IL-10, and IL-33, and others that appear to serve primarily anti-inflammatory functions, including IL-35 and IL-37. Here, we review the literature examining cytokines that regulate MC-mediated allergic immune responses.
Collapse
|
33
|
Kanagy WK, Cleyrat C, Fazel M, Lucero SR, Bruchez MP, Lidke KA, Wilson BS, Lidke DS. Docking of Syk to FcεRI is enhanced by Lyn but limited in duration by SHIP1. Mol Biol Cell 2022; 33:ar89. [PMID: 35793126 PMCID: PMC9582627 DOI: 10.1091/mbc.e21-12-0603] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Revised: 06/23/2022] [Accepted: 06/28/2022] [Indexed: 11/25/2022] Open
Abstract
The high-affinity immunoglobulin E (IgE) receptor, FcεRI, is the primary immune receptor found on mast cells and basophils. Signal initiation is classically attributed to phosphorylation of FcεRI β- and γ-subunits by the Src family kinase (SFK) Lyn, followed by the recruitment and activation of the tyrosine kinase Syk. FcεRI signaling is tuned by the balance between Syk-driven positive signaling and the engagement of inhibitory molecules, including SHIP1. Here, we investigate the mechanistic contributions of Lyn, Syk, and SHIP1 to the formation of the FcεRI signalosome. Using Lyn-deficient RBL-2H3 mast cells, we found that another SFK can weakly monophosphorylate the γ-subunit, yet Syk still binds the incompletely phosphorylated immunoreceptor tyrosine-based activation motifs (ITAMs). Once recruited, Syk further enhances γ-phosphorylation to propagate signaling. In contrast, the loss of SHIP1 recruitment indicates that Lyn is required for phosphorylation of the β-subunit. We demonstrate two noncanonical Syk binding modes, trans γ-bridging and direct β-binding, that can support signaling when SHIP1 is absent. Using single particle tracking, we reveal a novel role of SHIP1 in regulating Syk activity, where the presence of SHIP1 in the signaling complex acts to increase the Syk:receptor off-rate. These data suggest that the composition and dynamics of the signalosome modulate immunoreceptor signaling activities.
Collapse
Affiliation(s)
- William K. Kanagy
- Department of Pathology, University of New Mexico, Albuquerque, NM 87131
| | - Cédric Cleyrat
- Department of Pathology, University of New Mexico, Albuquerque, NM 87131
- Comprehensive Cancer Center, University of New Mexico, Albuquerque, NM 87131
| | - Mohamadreza Fazel
- Department of Physics, University of New Mexico, Albuquerque, NM 87131
| | - Shayna R. Lucero
- Department of Pathology, University of New Mexico, Albuquerque, NM 87131
| | - Marcel P. Bruchez
- Department of Biological Sciences and Department of Chemistry, Carnegie Mellon University, Pittsburgh, PA 15213
| | - Keith A. Lidke
- Comprehensive Cancer Center, University of New Mexico, Albuquerque, NM 87131
- Department of Physics, University of New Mexico, Albuquerque, NM 87131
| | - Bridget S. Wilson
- Department of Pathology, University of New Mexico, Albuquerque, NM 87131
- Comprehensive Cancer Center, University of New Mexico, Albuquerque, NM 87131
| | - Diane S. Lidke
- Department of Pathology, University of New Mexico, Albuquerque, NM 87131
- Comprehensive Cancer Center, University of New Mexico, Albuquerque, NM 87131
| |
Collapse
|
34
|
Hollstein MM, Matzke SS, Lorbeer L, Forkel S, Fuchs T, Lex C, Buhl T. Intracutaneous Skin Tests and Serum IgE Levels Cannot Predict the Grade of Anaphylaxis in Patients with Insect Venom Allergies. J Asthma Allergy 2022; 15:907-918. [PMID: 35836970 PMCID: PMC9274911 DOI: 10.2147/jaa.s367272] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Accepted: 06/23/2022] [Indexed: 11/29/2022] Open
Abstract
Background Allergies against Hymenoptera venoms are a major cause of severe anaphylaxis. Risk assessment for subjects with suspected allergy is difficult because there are currently no biomarkers that predict the likelihood of high-grade anaphylaxis other than several associated comorbidities. Objective We investigated the relationship between the severity of anaphylaxis and the results of intracutaneous skin tests (ICTs) together with serum levels of tryptase, total IgE, and venom-specific IgE, IgG, and IgG4. Methods We performed a retrospective evaluation of 194 patients who presented to a single medical center with allergies to bee venoms (Apis mellifera, Bombus spp.; n=24, 12.4%), vespid venoms (Vespula spp., Vespa spp., Polistes spp.; n=169, 87.1%), or both (n=1, 0.5%). Results Index bee stings occurred earlier in the year than vespid stings, although the latter were reported more frequently overall. On average, subjects who previously experienced grade IV anaphylaxis required higher dosages of venom to yield positive ICTs than those who exhibited lower grade responses. Patients diagnosed with grade IV anaphylaxis exhibited significantly lower levels of venom-specific IgE and IgG and trended toward elevated levels of tryptase. No significant differences in average levels of venom-specific IgG4 and total IgE were observed. Conclusion Our findings reveal that intracutaneous skin testing and levels of venom-specific IgE do not predict the degree of anaphylaxis that develops in patients with venom allergy. Furthermore, the month of the index sting is not a reliable means to differentiate bee from vespid stings in patients presenting with an uncertain history.
Collapse
Affiliation(s)
- Moritz M Hollstein
- Department of Dermatology, Venereology and Allergology, University Medical Centre Göttingen, Göttingen, Germany
| | - Silke S Matzke
- Department of Dermatology, Venereology and Allergology, University Medical Centre Göttingen, Göttingen, Germany
| | - Lisa Lorbeer
- Department of Dermatology, Venereology and Allergology, University Medical Centre Göttingen, Göttingen, Germany
| | - Susann Forkel
- Department of Dermatology, Venereology and Allergology, University Medical Centre Göttingen, Göttingen, Germany
| | - Thomas Fuchs
- Department of Dermatology, Venereology and Allergology, University Medical Centre Göttingen, Göttingen, Germany
| | - Christiane Lex
- Department of Pediatric Cardiology, Intensive Care Medicine and Neonatology with Pediatric Pneumology, University Medical Centre Göttingen, Göttingen, Germany
| | - Timo Buhl
- Department of Dermatology, Venereology and Allergology, University Medical Centre Göttingen, Göttingen, Germany
| |
Collapse
|
35
|
Buscail E, Deraison C. Postoperative Ileus: a Pharmacological Perspective. Br J Pharmacol 2022; 179:3283-3305. [PMID: 35048360 DOI: 10.1111/bph.15800] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Revised: 12/31/2021] [Accepted: 01/05/2022] [Indexed: 11/29/2022] Open
Abstract
Post-operative ileus (POI) is a frequent complication after abdominal surgery. The consequences of POI can be potentially serious such as bronchial inhalation or acute functional renal failure. Numerous advances in peri-operative management, particularly early rehabilitation, have made it possible to decrease POI. Despite this, the rate of prolonged POI ileus remains high and can be as high as 25% of patients in colorectal surgery. From a pathophysiological point of view, POI has two phases, an early neurological phase and a later inflammatory phase, to which we could add a "pharmacological" phase during which analgesic drugs, particularly opiates, play a central role. The aim of this review article is to describe the phases of the pathophysiology of POI, to analyse the pharmacological treatments currently available through published clinical trials and finally to discuss the different research areas for potential pharmacological targets.
Collapse
Affiliation(s)
- Etienne Buscail
- IRSD, INSERM, INRAE, ENVT, University of Toulouse, CHU Purpan (University Hospital Centre), Toulouse, France.,Department of digestive surgery, colorectal surgery unit, Toulouse University Hospital, Toulouse, France
| | - Céline Deraison
- IRSD, INSERM, INRAE, ENVT, University of Toulouse, CHU Purpan (University Hospital Centre), Toulouse, France
| |
Collapse
|
36
|
Wang Y, Ding Y, Li C, Gao J, Wang X, An H. Alpha-linolenic acid inhibits IgE-mediated anaphylaxis by inhibiting Lyn kinase and suppressing mast cell activation. Int Immunopharmacol 2021; 103:108449. [PMID: 34929479 DOI: 10.1016/j.intimp.2021.108449] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Revised: 11/28/2021] [Accepted: 12/05/2021] [Indexed: 12/11/2022]
Abstract
Excessive reactions to allergens can induce systemic, life-threatening physiological dysfunction (anaphylaxis) in humans. The surface of mast cells expresses high-affinity IgE receptors that play a vital role during anaphylaxis. Alpha-linolenic acid (ALA) is an essential non-toxic fatty acid in humans. Since it has been reported having potential to regulate pro-inflammatory reactions, we postulated that ALA could inhibit anaphylaxis by down-regulating Lyn kinase phosphorylation. We found that local and systematic inflammation induced by albumin from chicken egg white (OVA) were attenuated by ALA in vivo. Furthermore, ALA inhibited IgE-mediated Ca2+ mobilization, degranulation, and cytokine release in Laboratory of Allergic Disease 2 (LAD2) cells. The western blot results showed that ALA down-regulate the FcεRI/Lyn/Syk signaling pathway by suppressing Lyn kinase activity. Therefore, ALA could serve as a therapeutic drug candidate for preventing IgE-mediated anaphylaxis.
Collapse
Affiliation(s)
- Yuejin Wang
- Center for Translational Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China; College of Pharmacy, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| | - Yuanyuan Ding
- College of Pharmacy, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| | - Chaomei Li
- College of Pharmacy, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| | - Jiapan Gao
- College of Pharmacy, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| | - Xiaodong Wang
- Shaanxi Institute of Medical Device Quality Inspection, Xi'an, Shaanxi 712046, China
| | - Hongli An
- Center for Translational Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China; Key Laboratory for Tumor Precision Medicine of Shaanxi Province, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China.
| |
Collapse
|
37
|
Phull AR, Dhong KR, Park HJ. Lactic Acid Bacteria Fermented Cordyceps militaris (GRC-SC11) Suppresses IgE Mediated Mast Cell Activation and Type I Hypersensitive Allergic Murine Model. Nutrients 2021; 13:3849. [PMID: 34836105 PMCID: PMC8618942 DOI: 10.3390/nu13113849] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Revised: 10/24/2021] [Accepted: 10/27/2021] [Indexed: 01/16/2023] Open
Abstract
Cordyceps militaris (C. militaris) has various biomedical applications in traditional oriental medicine for different diseases including inflammatory and immune-dysregulated diseases. It is a reservoir of nutritional components such as cordycepin, polysaccharides, and antioxidants. To improve its bioactivity, we fermented C. militaris with a Pediococcus pentosaceus strain isolated from a salted small octopus (SC11). The current study aimed to evaluate whether P. pentosaceus (SC11) fermentation could enhance the anti-allergic potential of C. militaris cultured on germinated Rhynchosia nulubilis (GRC) against a type I hypersensitive reaction in in vitro and in vivo studies. Total antioxidant capacity and cordycepin content were significantly increased in GRC after SC11 fermentation. GRC-SC11 showed significantly enhanced anti-allergic responses by inhibiting immunoglobulin E (IgE)/antigen-induced degranulation in RBL-2H3 cells, compared to GRC. The results demonstrated the significant inhibition of phosphorylated spleen tyrosine kinase (Syk)/ p38/GRB2-associated binding protein 2 (Gab2)/c-jun in IgE/Ag-triggered RBL-2H3 cells. Furthermore, suppressed mRNA levels of interleukin-4 (IL-4) and tumor necrosis factor-α (TNF-α) in IgE/Ag-activated RBL-2H3 cells were observed. GRC-SC11 significantly ameliorated IgE-induced allergic reactions by suppressing the ear swelling, vascular permeability, and inflammatory cell infiltration in passive cutaneous anaphylaxis (PCA) BALB/c mice. In conclusion, GRC fermented with P.pentosaceus exerted enhanced anti-allergic effects, and increased the cordycepin content and antioxidants potential compared to GRC. It can be used as bio-functional food in the prevention and management of type I allergic diseases.
Collapse
Affiliation(s)
- Abdul-Rehman Phull
- Department of Food Science and Biotechnology, College of BioNano Technology, Gachon University, Seongnam 13120, Gyeonggi-do, Korea;
| | - Kyu-Ree Dhong
- Department of Life Science, College of BioNano Technology, Gachon University, Seongnam 13120, Gyeonggi-do, Korea;
| | - Hye-Jin Park
- Department of Food Science and Biotechnology, College of BioNano Technology, Gachon University, Seongnam 13120, Gyeonggi-do, Korea;
| |
Collapse
|
38
|
Tumor-Associated Mast Cells in Urothelial Bladder Cancer: Optimizing Immuno-Oncology. Biomedicines 2021; 9:biomedicines9111500. [PMID: 34829729 PMCID: PMC8614912 DOI: 10.3390/biomedicines9111500] [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: 09/20/2021] [Revised: 10/14/2021] [Accepted: 10/16/2021] [Indexed: 12/28/2022] Open
Abstract
Urothelial bladder cancer (UBC) is one of the most prevalent and aggressive malignancies. Recent evidence indicates that the tumor microenvironment (TME), including a variety of immune cells, is a critical modulator of tumor initiation, progression, evolution, and treatment resistance. Mast cells (MCs) in UBC are possibly involved in tumor angiogenesis, tissue remodeling, and immunomodulation. Moreover, tumor-infiltration by MCs has been reported in early-stage UBC patients. This infiltration is linked with a favorable or unfavorable prognosis depending on the tumor type and location. Despite the discrepancy of MC function in tumor progression, MCs can modify the TME to regulate the immunity and infiltration of tumors by producing an array of mediators. Nonetheless, the precise role of MCs in UBC tumor progression and evolution remains unknown. Thus, this review discusses some critical roles of MCs in UBC. Patients with UBC are treated at both early and late stages by immunotherapeutic methods, including intravenous bacillus Calmette–Guérin instillation and immune checkpoint blockade. An understanding of the patient response and resistance mechanisms in UBC is required to unlock the complete potential of immunotherapy. Since MCs are pivotal to understand the underlying processes and predictors of therapeutic responses in UBC, our review also focuses on possible immunotherapeutic treatments that involve MCs.
Collapse
|
39
|
Dhakal H, Kim MJ, Lee S, Choi YA, Kim N, Kwon TK, Khang D, Kim SH. Ursolic acid inhibits FcεRI-mediated mast cell activation and allergic inflammation. Int Immunopharmacol 2021; 99:107994. [PMID: 34435583 DOI: 10.1016/j.intimp.2021.107994] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Revised: 07/14/2021] [Accepted: 07/14/2021] [Indexed: 12/14/2022]
Abstract
BACKGROUND Mast cells are the primary cells that play a crucial role in the allergic diseases via secretion of diverse allergic mediators. Ursolic acid (UA) is a naturally occurring anti-inflammatory triterpenoid possessing various biological properties such as immune regulation, antioxidant, and anti-fibrotic. The aim of this study was to evaluate the effects of UA in FcεRI-mediated mast cell activation and allergic inflammation. METHODS In this study, mast cells were stimulated with immunoglobulin E (IgE) and the anti-allergic effects of UA were assessed by measuring the levels of allergic mediators. In vivo effects of UA were observed by generating passive cutaneous anaphylaxis (PCA) and active systemic anaphylaxis (ASA) in mouse model. RESULTS We found that UA inhibited the degranulation of mast cell by suppressing the intracellular calcium level in a concentration-dependent manner. UA inhibited the expression and the release of pro-inflammatory cytokines in mast cells. Anti-allergic effects of UA were demonstrated via suppression of FcεRI-mediated signaling molecules. In addition, UA inhibited the IgE-mediated PCA and ovalbumin-induced ASA reactions in a dose-dependent manner. CONCLUSIONS Based on these findings, we suggest that UA might have potential as a therapeutic candidate for the treatment of allergic inflammatory diseases via inhibition of FcεRI-mediated mast cell activation.
Collapse
Affiliation(s)
- Hima Dhakal
- Cell & Matrix Research Institute, Department of Pharmacology, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
| | - Min-Jong Kim
- Cell & Matrix Research Institute, Department of Pharmacology, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
| | - Soyoung Lee
- Immunoregulatory Materials Research Center, Korea Research Institute of Bioscience and Biotechnology, Jeongeup, Republic of Korea
| | - Young-Ae Choi
- Cell & Matrix Research Institute, Department of Pharmacology, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
| | - Namkyung Kim
- Cell & Matrix Research Institute, Department of Pharmacology, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
| | - Taeg Kyu Kwon
- Department of Immunology, School of Medicine, Keimyung University, Daegu, Republic of Korea
| | - Dongwoo Khang
- Department of Physiology, School of Medicine, Gachon University, Incheon, Republic of Korea.
| | - Sang-Hyun Kim
- Cell & Matrix Research Institute, Department of Pharmacology, School of Medicine, Kyungpook National University, Daegu, Republic of Korea.
| |
Collapse
|
40
|
Méndez-Enríquez E, Salomonsson M, Eriksson J, Janson C, Malinovschi A, Sellin ME, Hallgren J. IgE cross-linking induces activation of human and mouse mast cell progenitors. J Allergy Clin Immunol 2021; 149:1458-1463. [PMID: 34492259 DOI: 10.1016/j.jaci.2021.08.019] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Revised: 06/28/2021] [Accepted: 08/30/2021] [Indexed: 12/16/2022]
Abstract
BACKGROUND The concept of innate and adaptive effector cells that are repleted by maturing inert progenitor cell populations is changing. Mast cells develop from rare mast cell progenitors populating peripheral tissues at homeostatic conditions, or as a result of induced recruitment during inflammatory conditions. OBJECTIVE Because FcεRI-expressing mast cell progenitors are the dominating mast cell type during acute allergic lung inflammation in vivo, we hypothesized that they are activated by IgE cross-linking. METHODS Mouse peritoneal and human peripheral blood cells were sensitized and stimulated with antigen, or stimulated with anti-IgE, and the mast cell progenitor population analyzed for signs of activation by flow cytometry. Isolated peritoneal mast cell progenitors were studied before and after anti-IgE stimulation at single-cell level by time-lapse fluorescence microscopy. Lung mast cell progenitors were analyzed for their ability to produce IL-13 by intracellular flow cytometry in a mouse model of ovalbumin-induced allergic airway inflammation. RESULTS Sensitized mouse peritoneal mast cell progenitors demonstrate increased levels of phosphorylation of tyrosines on intracellular proteins (total tyrosine phosphorylation), and spleen tyrosine kinase (Syk) phosphorylation after antigen exposure. Anti-IgE induced cell surface-associated lysomal-associated membrane protein-1 (LAMP-1) in naive mast cell progenitors, and prompted loss of fluorescence signal and altered morphology of isolated cells loaded with lysotracker. In human mast cell progenitors, anti-IgE increased total tyrosine phosphorylation, cell surface-associated LAMP-1, and CD63. Lung mast cell progenitors from mice with ovalbumin-induced allergic airway inflammation produce IL-13. CONCLUSIONS Mast cell progenitors become activated by IgE cross-linking and may contribute to the pathology associated with acute allergic airway inflammation.
Collapse
Affiliation(s)
- Erika Méndez-Enríquez
- Department of Medical Biochemistry and Microbiology, BMC, Uppsala University, Uppsala, Sweden
| | - Maya Salomonsson
- Department of Medical Biochemistry and Microbiology, BMC, Uppsala University, Uppsala, Sweden
| | - Jens Eriksson
- Science for Life Laboratory, Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala, Sweden
| | - Christer Janson
- Department of Medical Sciences: Respiratory, Allergy and Sleep Research, Uppsala University, Uppsala, Sweden
| | - Andrei Malinovschi
- Department of Medical Sciences: Clinical Physiology, Uppsala University, Uppsala, Sweden
| | - Mikael E Sellin
- Science for Life Laboratory, Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala, Sweden
| | - Jenny Hallgren
- Department of Medical Biochemistry and Microbiology, BMC, Uppsala University, Uppsala, Sweden.
| |
Collapse
|
41
|
Park SJ, Sim KH, Shrestha P, Yang JH, Lee YJ. Perfluorooctane sulfonate and bisphenol A induce a similar level of mast cell activation via a common signaling pathway, Fyn-Lyn-Syk activation. Food Chem Toxicol 2021; 156:112478. [PMID: 34363875 DOI: 10.1016/j.fct.2021.112478] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Revised: 07/31/2021] [Accepted: 08/03/2021] [Indexed: 02/04/2023]
Abstract
Perfluoroalkyl compounds (PFCs) as food contaminants are widely distributed persistent organic pollutants (POPs) and have been suggested to induce immune dysfunction. However, their effects on immune function are not conclusive. Mast cells play a central role in allergic and non-allergic inflammatory responses. Therefore, we have examined the effects of PFCs (PFHxS, PFOA, PFOS) on mast cell-mediated inflammatory responses using in vitro mouse bone marrow-derived mast cells (BMMCs) and human mast cells (HMC-1) and in vivo mice model. The effects of PFCs were compared with those of bisphenol A (BPA), a well-studied environmental pollutant. Among PFCs tested, PFOS had the highest effects. Both PFOS and BPA increased degranulation and production of inflammatory eicosanoids in mast cells at a similar level, which subsequently led to increased skin edema and serum LTC4 and PGD2 in mice. Both PFOS and BPA increased not only downstream signaling (PLCγ1, AKT, ERK), but also upstream signaling (Fyn, Lyn, Syk/LAT) in mast cells. Taken together, PFOS and BPA induce mast cell-mediated inflammatory responses via a common signaling pathways. Our results may help establish the scientific basis for understanding the etiology of mast cell-mediated inflammatory responses and improve the immune dysfunction risk assessment for emerging POPs such as PFCs.
Collapse
Affiliation(s)
- Sung-Joon Park
- Department of Pharmacology/Toxicology, School of Medicine, Catholic University of Daegu, Daegu, Republic of Korea
| | - Kyeong Hwa Sim
- Department of Pharmacology/Toxicology, School of Medicine, Catholic University of Daegu, Daegu, Republic of Korea
| | - Prafulla Shrestha
- Department of Pharmacology/Toxicology, School of Medicine, Catholic University of Daegu, Daegu, Republic of Korea
| | - Jae-Ho Yang
- Department of Pharmacology/Toxicology, School of Medicine, Catholic University of Daegu, Daegu, Republic of Korea
| | - Youn Ju Lee
- Department of Pharmacology/Toxicology, School of Medicine, Catholic University of Daegu, Daegu, Republic of Korea.
| |
Collapse
|
42
|
Wang Z, Franke K, Zuberbier T, Babina M. Cytokine Stimulation via MRGPRX2 Occurs with Lower Potency than by FcεRI-aggregation but with Similar Dependence on the ERK1/2 Module in Human Skin Mast Cells. J Invest Dermatol 2021; 142:414-424.e8. [PMID: 34329659 DOI: 10.1016/j.jid.2021.07.153] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Revised: 07/04/2021] [Accepted: 07/06/2021] [Indexed: 12/17/2022]
Abstract
Skin mast cells (MCs) contribute to chronic dermatoses that partially rely on MC-derived cytokines. The discovery of MRGPRX2 explains MC-dependent symptoms independently of FcεRI-activation. Here, we investigated whether MRGPRX2 can elicit cytokines, determined its relative potency versus FcεRI and addressed the underlying mechanisms. MRGPRX2-activation by compound 48/80 or Substance P on skin MCs induced TNF-α, IL-8, IL-13, CCL1, CCL2 mRNA and protein, yet induction was typically reduced compared with FcεRI-crosslinking. Generally, cytokine secretion required de-novo-synthesis with maximum accumulation at ≈8 h. Addressing key kinases revealed robust, rapid (1 min), and lasting (30 min) phosphorylation of ERK1/2 following MRGPRX2-ligation, while pp38, and pAKT signals were weaker, and pJNK hardly detectable. The kinase spectrum following FcεRI-aggregation was comparable, but responses considerably delayed. The MEK/ERK pathway was essential for all cytokines examined and four inhibitors of this module gave complete suppression. Variable and weaker contribution was found for PI3K>JNK>p38. Strikingly, cytokine profiles and signaling prerequisites were similar for MRGPRX2 and FcεRI and likely mainly dictated by the MC subset. Collectively, in skin MCs, the physiological producers of MRGPRX2, agonist binding elicits cytokines, yet less efficiently than FcεRI-aggregation. MRGPRX2-associated inflammation may thus be less tissue-destructive than responses to allergic challenge.
Collapse
Affiliation(s)
- Zhao Wang
- Department of Dermatology and Allergy, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany; Department of Dermatology, The Second Affiliated Hospital, Northwest Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Kristin Franke
- Department of Dermatology and Allergy, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Torsten Zuberbier
- Department of Dermatology and Allergy, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Magda Babina
- Department of Dermatology and Allergy, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany.
| |
Collapse
|
43
|
Jiménez M, Cervantes-García D, Córdova-Dávalos LE, Pérez-Rodríguez MJ, Gonzalez-Espinosa C, Salinas E. Responses of Mast Cells to Pathogens: Beneficial and Detrimental Roles. Front Immunol 2021; 12:685865. [PMID: 34211473 PMCID: PMC8240065 DOI: 10.3389/fimmu.2021.685865] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Accepted: 05/28/2021] [Indexed: 12/19/2022] Open
Abstract
Mast cells (MCs) are strategically located in tissues close to the external environment, being one of the first immune cells to interact with invading pathogens. They are long living effector cells equipped with different receptors that allow microbial recognition. Once activated, MCs release numerous biologically active mediators in the site of pathogen contact, which induce vascular endothelium modification, inflammation development and extracellular matrix remodeling. Efficient and direct antimicrobial mechanisms of MCs involve phagocytosis with oxidative and non-oxidative microbial destruction, extracellular trap formation, and the release of antimicrobial substances. MCs also contribute to host defense through the attraction and activation of phagocytic and inflammatory cells, shaping the innate and adaptive immune responses. However, as part of their response to pathogens and under an impaired, sustained, or systemic activation, MCs may contribute to tissue damage. This review will focus on the current knowledge about direct and indirect contribution of MCs to pathogen clearance. Antimicrobial mechanisms of MCs are addressed with special attention to signaling pathways involved and molecular weapons implicated. The role of MCs in a dysregulated host response that can increase morbidity and mortality is also reviewed and discussed, highlighting the complexity of MCs biology in the context of host-pathogen interactions.
Collapse
Affiliation(s)
- Mariela Jiménez
- Laboratory of Immunology, Department of Microbiology, Universidad Autónoma de Aguascalientes, Aguascalientes, Mexico
| | - Daniel Cervantes-García
- Laboratory of Immunology, Department of Microbiology, Universidad Autónoma de Aguascalientes, Aguascalientes, Mexico.,Cátedras CONACYT, National Council of Science and Technology, Mexico City, Mexico
| | - Laura E Córdova-Dávalos
- Laboratory of Immunology, Department of Microbiology, Universidad Autónoma de Aguascalientes, Aguascalientes, Mexico
| | - Marian Jesabel Pérez-Rodríguez
- Department of Pharmacobiology, Centro de Investigación y de Estudios Avanzados (Cinvestav), Unidad Sede Sur, Mexico City, Mexico
| | - Claudia Gonzalez-Espinosa
- Department of Pharmacobiology, Centro de Investigación y de Estudios Avanzados (Cinvestav), Unidad Sede Sur, Mexico City, Mexico
| | - Eva Salinas
- Laboratory of Immunology, Department of Microbiology, Universidad Autónoma de Aguascalientes, Aguascalientes, Mexico
| |
Collapse
|
44
|
The pLysRS-Ap 4A Pathway in Mast Cells Regulates the Switch from Host Defense to a Pathological State. Int J Mol Sci 2021; 22:ijms22115620. [PMID: 34070694 PMCID: PMC8198065 DOI: 10.3390/ijms22115620] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2021] [Revised: 05/22/2021] [Accepted: 05/23/2021] [Indexed: 11/17/2022] Open
Abstract
The innate and adaptive immune systems play an essential role in host defense against pathogens. Various signal transduction pathways monitor and balance the immune system since an imbalance may promote pathological states such as allergy, inflammation, and cancer. Mast cells have a central role in the regulation of the innate/adaptive immune system and are involved in the pathogenesis of many inflammatory and allergic diseases by releasing inflammatory mediators such as histamines, proteases, chemotactic factors, and cytokines. Although various signaling pathways are associated with mast cell activation, our discovery and characterization of the pLysRS-Ap4A signaling pathway in these cells provided an additional important step towards a full understanding of the intracellular mechanisms involved in mast cell activation. In the present review, we will discuss in depth this signaling pathway’s contribution to host defense and the pathological state.
Collapse
|
45
|
Lichterman JN, Reddy SM. Mast Cells: A New Frontier for Cancer Immunotherapy. Cells 2021; 10:cells10061270. [PMID: 34063789 PMCID: PMC8223777 DOI: 10.3390/cells10061270] [Citation(s) in RCA: 72] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2021] [Revised: 05/13/2021] [Accepted: 05/19/2021] [Indexed: 12/24/2022] Open
Abstract
Mast cells are unique tissue-resident immune cells of the myeloid lineage that have long been implicated in the pathogenesis of allergic and autoimmune disorders. More recently, mast cells have been recognized as key orchestrators of anti-tumor immunity, modulators of the cancer stroma, and have also been implicated in cancer cell intrinsic properties. As such, mast cells are an underrecognized but very promising target for cancer immunotherapy. In this review, we discuss the role of mast cells in shaping cancer and its microenvironment, the interaction between mast cells and cancer therapies, and strategies to target mast cells to improve cancer outcomes. Specifically, we address (1) decreasing cell numbers through c-KIT inhibition, (2) modulating mast cell activation and phenotype (through mast cell stabilizers, FcεR1 signaling pathway activators/inhibitors, antibodies targeting inhibitory receptors and ligands, toll like receptor agonists), and (3) altering secreted mast cell mediators and their downstream effects. Finally, we discuss the importance of translational research using patient samples to advance the field of mast cell targeting to optimally improve patient outcomes. As we aim to expand the successes of existing cancer immunotherapies, focused clinical and translational studies targeting mast cells in different cancer contexts are now warranted.
Collapse
Affiliation(s)
- Jake N. Lichterman
- Division of Hematology/Oncology, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA;
| | - Sangeetha M. Reddy
- Division of Hematology/Oncology, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA;
- Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
- Correspondence: ; Tel.: +1-214-648-4180
| |
Collapse
|
46
|
Filho EGF, da Silva EZM, Ong HL, Swaim WD, Ambudkar IS, Oliver C, Jamur MC. RACK1 plays a critical role in mast cell secretion and Ca2+ mobilization by modulating F-actin dynamics. J Cell Sci 2021; 134:263932. [PMID: 34550354 DOI: 10.1242/jcs.252585] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Accepted: 03/15/2021] [Indexed: 11/20/2022] Open
Abstract
Although RACK1 is known to act as a signaling hub in immune cells, its presence and role in mast cells (MCs) is undetermined. MC activation via antigen stimulation results in mediator release and is preceded by cytoskeleton reorganization and Ca2+ mobilization. In this study, we found that RACK1 was distributed throughout the MC cytoplasm both in vivo and in vitro. After RACK1 knockdown (KD), MCs were rounded, and the cortical F-actin was fragmented. Following antigen stimulation, in RACK1 KD MCs, there was a reduction in cortical F-actin, an increase in monomeric G-actin and a failure to organize F-actin. RACK1 KD also increased and accelerated degranulation. CD63+ secretory granules were localized in F-actin-free cortical regions in non-stimulated RACK1 KD MCs. Additionally, RACK1 KD increased antigen-stimulated Ca2+ mobilization, but attenuated antigen-stimulated depletion of ER Ca2+ stores and thapsigargin-induced Ca2+ entry. Following MC activation there was also an increase in interaction of RACK1 with Orai1 Ca2+-channels, β-actin and the actin-binding proteins vinculin and MyoVa. These results show that RACK1 is a critical regulator of actin dynamics, affecting mediator secretion and Ca2+ signaling in MCs. This article has an associated First Person interview with the first author of the paper.
Collapse
Affiliation(s)
- Edismauro G Freitas Filho
- Department of Cell and Molecular Biology and Pathogenic Bioagents, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Av. Bandeirantes 3900, Ribeirão Preto, SP 14049-900, Brazil
| | - Elaine Z M da Silva
- Department of Cell and Molecular Biology and Pathogenic Bioagents, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Av. Bandeirantes 3900, Ribeirão Preto, SP 14049-900, Brazil
| | - Hwei Ling Ong
- Secretory Physiology Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD 20892, USA
| | - William D Swaim
- Secretory Physiology Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD 20892, USA
| | - Indu S Ambudkar
- Secretory Physiology Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD 20892, USA
| | - Constance Oliver
- Department of Cell and Molecular Biology and Pathogenic Bioagents, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Av. Bandeirantes 3900, Ribeirão Preto, SP 14049-900, Brazil
| | - Maria Célia Jamur
- Department of Cell and Molecular Biology and Pathogenic Bioagents, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Av. Bandeirantes 3900, Ribeirão Preto, SP 14049-900, Brazil
| |
Collapse
|
47
|
Blank U, Huang H, Kawakami T. The high affinity IgE receptor: a signaling update. Curr Opin Immunol 2021; 72:51-58. [PMID: 33838574 DOI: 10.1016/j.coi.2021.03.015] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Accepted: 03/22/2021] [Indexed: 01/01/2023]
Abstract
Here we update receptor proximal and distant signaling events of the mast cell high affinity IgE receptor (FcεRI) launching immediate type I hypersensitivity and an inflammatory cytokine-chemokine cascade. Different physiologic antigen concentrations, their affinity, and valency for the IgE ligand produce distinct intracellular signaling events with different outcomes. Investigating mast cell degranulation has revealed a complex molecular machinery that relays proximal signaling to cytoskeletal reorganization, granule transport and membrane fusion. Several new phosphorylation- and calcium-responsive effectors have been described. FcεRI signaling also promotes de novo gene transcription. Recent progress has identified enhancers at genes that are upregulated in mast cells after stimulation through FcεRI using next generation sequencing methods. Enhancers at genes that respond to antigenic stimulation in human mast cells revealed Ca2+-dependency. Stimulation-responsive super enhancers in mouse mast cells have also been identified. Mast cell lineage-determining transcription factor GATA2 primes these enhancers to respond to antigenic stimulation.
Collapse
Affiliation(s)
- Ulrich Blank
- Université de Paris, Centre de Recherche sur l'Inflammation, INSERM UMR1149, CNRS ERL8252, Faculté de Médecine site Bichat, Paris, France; Laboratoire d'Excellence Inflamex, Paris, France.
| | - Hua Huang
- Department of Immunology and Genomic Medicine, National Jewish Health, Denver, CO 80206, USA; Department of Immunology and Microbiology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Toshiaki Kawakami
- Laboratory of Allergic Diseases, Center for Autoimmunity and Inflammation, La Jolla Institute for Immunology, La Jolla, CA 92037, USA; Department of Dermatology, School of Medicine, University of California San Diego, La Jolla, CA 92093, USA
| |
Collapse
|
48
|
Zorig A, Toko R, Sukhbold E, Takasugi M, Arai H. Echinacea purpurea water extracts suppress the release of chemical mediators from mast cells. Biosci Biotechnol Biochem 2021; 85:931-940. [PMID: 33686410 DOI: 10.1093/bbb/zbaa125] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Accepted: 12/23/2020] [Indexed: 11/12/2022]
Abstract
Histamine and leukotrienes (LTs), the chemical mediators released from mast cells, play an important role in type-I allergies such as hay fever. Echinacea purpurea (EP) has traditionally been used for herbal tea and has been reported to show biological functions. We evaluated the inhibitory activity of water extracts of EP petals, leaves, and stems against the chemical mediators released from mast cell lines. Petal and leaf extracts exhibited a significant inhibitory effect on histamine release from the stimulated cells, while the stem extract did not exert any effect. Activity of the petal extract was much stronger than that of the leaf extract. All the extracts significantly suppressed LTB4 production in the stimulated cells and displayed similar activities. The petal extract decreased Syk phosphorylation and Ca2+ influx associated with signal transduction in the stimulated cells. These results suggest that EP petal extract may have a relieving effect on allergic symptoms.
Collapse
Affiliation(s)
- Anuu Zorig
- Department of Biotechnology and Environmental Chemistry, Kitami Institute of Technology, Kitami, Hokkaido, Japan
| | - Rine Toko
- Department of Biotechnology and Environmental Chemistry, Kitami Institute of Technology, Kitami, Hokkaido, Japan
| | - Enkhtsetseg Sukhbold
- Department of Biotechnology and Environmental Chemistry, Kitami Institute of Technology, Kitami, Hokkaido, Japan
| | - Mikako Takasugi
- Department of Life Science, Kyushu Sangyo University, Higashi-ku, Fukuoka, Japan
| | - Hirofumi Arai
- Department of Biotechnology and Environmental Chemistry, Kitami Institute of Technology, Kitami, Hokkaido, Japan
| |
Collapse
|
49
|
Strategies for Mast Cell Inhibition in Food Allergy. THE YALE JOURNAL OF BIOLOGY AND MEDICINE 2020; 93:719-731. [PMID: 33380934 PMCID: PMC7757070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Mast cells are tissue resident allergic effector cells that drive IgE-mediated food allergies. There are several steps leading to mast cell activation in the context of allergic disease that can be targeted to prevent mast cell activation and degranulation. These include blocking IgE-FcεRI crosslinking and type 2 cytokine receptor activation; modulating cell-surface neural chemical receptors; stabilizing mast cell membranes to prevent co-localization of activating receptors; impeding intracellular signaling; and engaging cell surface inhibitory receptors. This review highlights several ITIM-containing inhibitory mast cell surface receptors that could serve as pharmaceutical targets to prevent mast cell activation and degranulation in the context of food allergy. When activated, these ITIM-containing inhibitory receptors recruit the phosphatases SHP-1, SHP-2, and/or SHIP to dephosphorylate the tyrosine kinases responsible for activation signals downstream of the IgE-FcεRI complex. We describe several members of the Ig and Ig-like inhibitory receptor and C-type lectin inhibitory receptor superfamilies. Fundamental studies exploring the behavior of these receptors within the context of experimental food allergy models are needed. A deeper understanding of how these receptors modulate mast cell-driven food allergic responses will shape future strategies to harness these inhibitory receptors to treat food allergy.
Collapse
|
50
|
Wang X, Ilarraza R, Tancowny BP, Alam SB, Kulka M. Disrupted Lipid Raft Shuttling of FcεRI by n-3 Polyunsaturated Fatty Acid Is Associated With Ligation of G Protein-Coupled Receptor 120 (GPR120) in Human Mast Cell Line LAD2. Front Nutr 2020; 7:597809. [PMID: 33330598 PMCID: PMC7732685 DOI: 10.3389/fnut.2020.597809] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2020] [Accepted: 10/20/2020] [Indexed: 12/26/2022] Open
Abstract
n-3 polyunsaturated fatty acids (PUFA) influences a variety of disease conditions, such as hypertension, heart disease, diabetes, cancer and allergic diseases, by modulating membrane constitution, inhibiting production of proinflammatory eicosanoids and cytokines, and binding to cell surface and nuclear receptors. We have previously shown that n-3 PUFA inhibit mast cell functions by disrupting high affinity IgE receptor (FcεRI) lipid raft partitioning and subsequent suppression of FcεRI signaling in mouse bone marrow-derived mast cells. However, it is still largely unknown how n-3 PUFA modulate human mast cell function, which could be attributed to multiple mechanisms. Using a human mast cell line (LAD2), we have shown similar modulating effects of n-3 PUFA on FcεRI lipid raft shuttling, FcεRI signaling, and mediator release after cell activation through FcεRI. We have further shown that these effects are at least partially associated with ligation of G protein-coupled receptor 120 expressed on LAD2 cells. This observation has advanced our mechanistic knowledge of n-3 PUFA's effect on mast cells and demonstrated the interplay between n-3 PUFA, lipid rafts, FcεRI, and G protein-coupled receptor 120. Future research in this direction may present new targets for nutritional intervention and therapeutic agents.
Collapse
Affiliation(s)
- Xiaofeng Wang
- Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, AB, Canada
| | - Ramses Ilarraza
- Department of Medical Microbiology and Immunology, University of Alberta, Edmonton, AB, Canada
| | - Brian P Tancowny
- Department of Biochemistry, Prion Research Institute, University of Alberta, Edmonton, AB, Canada
| | - Syed Benazir Alam
- Department of Medical Microbiology and Immunology, University of Alberta, Edmonton, AB, Canada.,National Research Council Canada, Nanotechnology Research Centre, Edmonton, AB, Canada
| | - Marianna Kulka
- Department of Medical Microbiology and Immunology, University of Alberta, Edmonton, AB, Canada.,National Research Council Canada, Nanotechnology Research Centre, Edmonton, AB, Canada
| |
Collapse
|